1
|
Knabl J, Ye Y, Desoye G, Jeschke U. HLA-G - evolvement from a trophoblast specific marker to a checkpoint molecule in cancer, a narrative review about the specific role in breast- and gynecological cancer. J Reprod Immunol 2024; 166:104385. [PMID: 39432974 DOI: 10.1016/j.jri.2024.104385] [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: 04/08/2024] [Revised: 10/07/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024]
Abstract
Human leukocyte antigen G (HLA-G) is known as a non-classical molecule of the major histocompatibility complex class Ib and downregulates the mother's immune response against the fetus during pregnancy, thereby generating immune tolerance. Due to the latter effect, HLA-G is also referred to as an immune checkpoint molecule. Originally identified on extravillous trophoblasts, HLA-G is already known to induce immune tolerance at various stages of the immune response, for example through cell differentiation and proliferation, cytolysis and cytokine secretion. Because of these functions, HLA-G is involved in various processes of cancer progression, but a comprehensive review of the role of HLA-G in gynecologic cancers is lacking. Therefore, this review focuses on the existing knowledge of HLA-G in ovarian cancer, endometrial cancer, cervical cancer and breast cancer. HLA-G is predominantly expressed in cancer tissues adjacent to the extravillous trophoblast. Therefore, modulating its expression in the cancer target tissues of cancer patients could be a potential therapeutic approach to treat these diseases.
Collapse
Affiliation(s)
- Julia Knabl
- Department of Gynecology and Obstetrics, University Hospital, LMU Munich, Marchioninistr.15, Munich 81377 , Germany; Department of Obstetrics, Klinik Hallerwiese, St.-Johannis Mühlgasse 19, Nürnberg 90419, Germany
| | - Yao Ye
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University School of Medicine, Nanjing, China
| | - Gernot Desoye
- Department of Obstetrics and Gynecology, Medical University Graz, Auenbruggerplatz 14, Graz A-8036, Austria
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstr. 2, Augsburg 86156, Germany.
| |
Collapse
|
2
|
Zhou Y, Qin Y, Sun C, Liu K, Zhang W, Găman MA, Chen Y. Cell-bound membrane vesicles contain antioxidative proteins and probably have an antioxidative function in cells or a therapeutic potential. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
3
|
Jiang H, Thapa P, Hao Y, Ding N, Alshahrani A, Wei Q. Protein Disulfide Isomerases Function as the Missing Link Between Diabetes and Cancer. Antioxid Redox Signal 2022; 37:1191-1205. [PMID: 36000195 PMCID: PMC9805878 DOI: 10.1089/ars.2022.0098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/11/2022] [Indexed: 01/13/2023]
Abstract
Significance: Diabetes has long been recognized as an independent risk factor for cancer, but there is insufficient mechanistic understanding of biological mediators that bridge two disorders together. Understanding the pathogenic association between diabetes and cancer has become the focus of many studies, and findings are potentially valuable for the development of effective preventive or therapeutic strategies for both disorders. Recent Advances: A summary of literature reveals a possible connection between diabetes and cancer through the family of protein disulfide isomerase (PDI). Historical as well as the most recent findings on the structure, biochemistry, and biology of the PDI family were summarized in this review. Critical Issues: PDIs in general function as redox enzymes and protein chaperones to control the quality of proteins by correcting or otherwise eliminating misfolded proteins in conditions of oxidative stress and endoplasmic reticulum stress, respectively. However, individual members of the PDI family may contribute uniquely to the pathogenesis of diabetes and cancer. Studies of exemplary members such as protein disulfide isomerase-associated (PDIA) 1, PDIA6, and PDIA15 were reviewed to highlight their contributions in the pathogenesis of diabetes and cancer and how they can be potential links bridging the two disorders through the cross talk of signaling pathways. Future Directions: Apparently ubiquitous presence of the PDIs creates difficulties and challenges for scientific community to develop targeted therapeutics for the treatment of diabetes and cancer simultaneously. Understanding molecular contribution of individual PDI in the context of specific disease may provide some insights into the development of mechanism-based target-directed therapeutics. Antioxid. Redox Signal. 37, 1191-1205.
Collapse
Affiliation(s)
- Hong Jiang
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Pratik Thapa
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Yanning Hao
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Na Ding
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Aziza Alshahrani
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Qiou Wei
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, Kentucky, USA
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| |
Collapse
|
4
|
Serrano-Quintero A, Sequeda-Juárez A, Pérez-Hernández CA, Sosa-Delgado SM, Mendez-Tenorio A, Ramón-Gallegos E. Immunogenic analysis of epitope-based vaccine candidate induced by photodynamic therapy in MDA-MB-231 triple-negative breast cancer cells. Photodiagnosis Photodyn Ther 2022; 40:103174. [PMID: 36602069 DOI: 10.1016/j.pdpdt.2022.103174] [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: 08/05/2022] [Revised: 09/20/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Photodynamic therapy (PDT) is used to treat tumors through selective cytotoxic effects. PDT induces damage-associated molecular patterns (DAMPs) expression, which can cause an immunogenic death cell (IDC). In this study we identified potential immunogenic epitopes generated by PDT on triple-negative breast cancer cell line (MDA-MB-231). METHODS MDA-MB-231 cells were exposed to PDT using ALA (160 µg/mL)/630 nm at 8 J/cm2. Membrane proteins were extracted and separated by 2D PAGE. Proteins overexpressed were identified by LC-MS/MS and analyzed in silico through a peptide-HLA docking in order to identify the epitopes with more immunogenicity and antigenicity properties, as well as lower allergenicity and toxicity activity. The selected peptides were evaluated in response to macrophage activation and cytokine release by flow cytometry. RESULTS Differential proteins were overexpressed in the cells treated with PDT. A group of 16 peptides were identified from them, established in a rigorous selection by measuring antigenicity, immunogenicity, allergenicity, and toxicity in silico. The final selection was based on molecular dynamics, where 2 peptides showed the highest stability regarding to the RMSD value. These peptides were obtained from the proteins calreticulin and HSP90. The cytokine analysis evidenced macrophage activation by the releasing of TNF. CONCLUSION Two peptides were identified from calreticulin and HSP90; proteins induced by PDT in MDA-MB-231 cells. Both epitopes showed immunogenic potential as a peptide-based vaccine for triple-negative breast cancer.
Collapse
Affiliation(s)
- Alina Serrano-Quintero
- Laboratorio de Citopatología Ambiental, ENCB, Instituto Politécnico Nacional (IPN), Campus Zacatenco, Calle Wilfrido Massieu Esquina Cda. Manuel Stampa, Col. Zacatenco. Alcaldia Gustavo A. Madero, Mexico City C.P. 07738, Mexico
| | - Alfonso Sequeda-Juárez
- Laboratorio de Citopatología Ambiental, ENCB, Instituto Politécnico Nacional (IPN), Campus Zacatenco, Calle Wilfrido Massieu Esquina Cda. Manuel Stampa, Col. Zacatenco. Alcaldia Gustavo A. Madero, Mexico City C.P. 07738, Mexico
| | - C Angélica Pérez-Hernández
- Laboratorio de Citopatología Ambiental, ENCB, Instituto Politécnico Nacional (IPN), Campus Zacatenco, Calle Wilfrido Massieu Esquina Cda. Manuel Stampa, Col. Zacatenco. Alcaldia Gustavo A. Madero, Mexico City C.P. 07738, Mexico
| | - Sara M Sosa-Delgado
- Laboratorio de Citopatología Ambiental, ENCB, Instituto Politécnico Nacional (IPN), Campus Zacatenco, Calle Wilfrido Massieu Esquina Cda. Manuel Stampa, Col. Zacatenco. Alcaldia Gustavo A. Madero, Mexico City C.P. 07738, Mexico
| | - Alfonso Mendez-Tenorio
- Laboratorio de Bioinformática y Biotecnología Genómica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Eva Ramón-Gallegos
- Laboratorio de Citopatología Ambiental, ENCB, Instituto Politécnico Nacional (IPN), Campus Zacatenco, Calle Wilfrido Massieu Esquina Cda. Manuel Stampa, Col. Zacatenco. Alcaldia Gustavo A. Madero, Mexico City C.P. 07738, Mexico.
| |
Collapse
|
5
|
Jayathirtha M, Neagu AN, Whitham D, Alwine S, Darie CC. Investigation of the effects of overexpression of jumping translocation breakpoint (JTB) protein in MCF7 cells for potential use as a biomarker in breast cancer. Am J Cancer Res 2022; 12:1784-1823. [PMID: 35530281 PMCID: PMC9077082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/27/2022] [Indexed: 06/14/2023] Open
Abstract
Jumping translocation breakpoint (JTB) gene acts as a tumor suppressor or an oncogene in different malignancies, including breast cancer (BC), where it was reported as overexpressed. However, the molecular functions, biological processes and underlying mechanisms through which JTB protein causes increased cell growth, proliferation and invasion is still not fully deciphered. Our goal is to identify the functions of JTB protein by cellular proteomics approaches. MCF7 breast cancer cells were transfected with sense orientation of hJTB cDNA in HA, His and FLAG tagged CMV expression vector to overexpress hJTB and the expression levels were confirmed by Western blotting (WB). Proteins extracted from transfected cells were separated by SDS-PAGE and the in-gel digested peptides were analyzed by nano-liquid chromatography tandem mass spectrometry (nanoLC-MS/MS). By comparing the proteome of cells with upregulated conditions of JTB vs control and identifying the protein dysregulation patterns, we aim to understand the function of this protein and its contribution to tumorigenesis. Gene Set Enrichment Analysis (GSEA) algorithm was performed to investigate the biological processes and pathways that are associated with the JTB protein upregulation. The results demonstrated four significantly enriched gene sets from the following significantly upregulated pathways: mitotic spindle assembly, estrogen response late, epithelial-to-mesenchymal transition (EMT) and estrogen response early. JTB protein itself is involved in mitotic spindle pathway by its role in cell division/cytokinesis, and within estrogen response early and late pathways, contributing to discrimination between luminal and mesenchymal breast cancer. Thus, the overexpressed JTB condition was significantly associated with an increased expression of ACTNs, FLNA, FLNB, EZR, MYOF, COL3A1, COL11A1, HSPA1A, HSP90A, WDR, EPPK1, FASN and FOXA1 proteins related to deregulation of cytoskeletal organization and biogenesis, mitotic spindle organization, ECM remodeling, cellular response to estrogen, proliferation, migration, metastasis, increased lipid biogenesis, endocrine therapy resistance, antiapoptosis and discrimination between different breast cancer subtypes. Other upregulated proteins for overexpressed JTB condition are involved in multiple cellular functions and pathways that become dysregulated, such as tumor microenvironment (TME) acidification, the transmembrane transport pathways, glycolytic flux, iron metabolism and oxidative stress, metabolic reprogramming, nucleocytosolic mRNA transport, transcriptional activation, chromatin remodeling, modulation of cell death pathways, stress responsive pathways, and cancer drug resistance. The downregulated proteins for overexpressed JTB condition are involved in adaptive communication between external and internal environment of cells and maintenance between pro-apoptotic and anti-apoptotic signaling pathways, vesicle trafficking and secretion, DNA lesions repair and suppression of genes involved in tumor progression, proteostasis, redox state regulation, biosynthesis of macromolecules, lipolytic pathway, carbohydrate metabolism, dysregulation of ubiquitin-mediated degradation system, cancer cell immune escape, cell-to-cell and cell-to-ECM interactions, and cytoskeletal behaviour. There were no significantly enriched downregulated pathways.
Collapse
Affiliation(s)
- Madhuri Jayathirtha
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| | - Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of IasiCarol I Bvd. No. 22, Iasi 700505, Romania
| | - Danielle Whitham
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| | - Shelby Alwine
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| | - Costel C Darie
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| |
Collapse
|
6
|
Yang S, Jackson C, Karapetyan E, Dutta P, Kermah D, Wu Y, Wu Y, Schloss J, Vadgama JV. Roles of Protein Disulfide Isomerase in Breast Cancer. Cancers (Basel) 2022; 14:745. [PMID: 35159012 PMCID: PMC8833603 DOI: 10.3390/cancers14030745] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 02/08/2023] Open
Abstract
Protein disulfide isomerase (PDI) is the endoplasmic reticulum (ER)'s most abundant and essential enzyme and serves as the primary catalyst for protein folding. Due to its apparent role in supporting the rapid proliferation of cancer cells, the selective blockade of PDI results in apoptosis through sustained activation of UPR pathways. The functions of PDI, especially in cancers, have been extensively studied over a decade, and recent research has explored the use of PDI inhibitors in the treatment of cancers but with focus areas of other cancers, such as brain or ovarian cancer. In this review, we discuss the roles of PDI members in breast cancer and PDI inhibitors used in breast cancer research. Additionally, a few PDI members may be suggested as potential molecular targets for highly metastatic breast cancers, such as TNBC, that require more attention in future research.
Collapse
Affiliation(s)
- Suhui Yang
- Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA; (E.K.); (P.D.); (Y.W.); (Y.W.); (J.S.)
- School of Pharmacy, American University of Health Sciences, Signal Hill, CA 90755, USA
| | - Chanel Jackson
- Post Baccalaureate Pre-Medical Program, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA;
| | - Eduard Karapetyan
- Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA; (E.K.); (P.D.); (Y.W.); (Y.W.); (J.S.)
| | - Pranabananda Dutta
- Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA; (E.K.); (P.D.); (Y.W.); (Y.W.); (J.S.)
| | - Dulcie Kermah
- Urban Health Institute, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA;
| | - Yong Wu
- Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA; (E.K.); (P.D.); (Y.W.); (Y.W.); (J.S.)
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, The University of California at Los Angeles, Los Angeles, CA 90059, USA
| | - Yanyuan Wu
- Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA; (E.K.); (P.D.); (Y.W.); (Y.W.); (J.S.)
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, The University of California at Los Angeles, Los Angeles, CA 90059, USA
| | - John Schloss
- Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA; (E.K.); (P.D.); (Y.W.); (Y.W.); (J.S.)
- School of Pharmacy, American University of Health Sciences, Signal Hill, CA 90755, USA
| | - Jaydutt V. Vadgama
- Division of Cancer Research and Training, Department of Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA; (E.K.); (P.D.); (Y.W.); (Y.W.); (J.S.)
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, The University of California at Los Angeles, Los Angeles, CA 90059, USA
| |
Collapse
|
7
|
Bakker EY, Fujii M, Krstic-Demonacos M, Demonacos C, Alhammad R. Protein disulfide isomerase A1‑associated pathways in the development of stratified breast cancer therapies. Int J Oncol 2022; 60:16. [PMID: 35014681 PMCID: PMC8776328 DOI: 10.3892/ijo.2022.5306] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/07/2021] [Indexed: 11/18/2022] Open
Abstract
The oxidoreductase protein disulfide isomerase A1 (PDIA1) functions as a cofactor for many transcription factors including estrogen receptor α (ERα), nuclear factor (NF)-κB, nuclear factor erythroid 2-like 2 (NRF2) and regulates the protein stability of the tumor suppressor p53. Taking this into account we hypothesized that PDIA1, by differentially modulating the gene expression of a diverse subset of genes in the ERα-positive vs. the ERα-negative breast cancer cells, might modify dissimilar pathways in the two types of breast cancer. This hypothesis was investigated using RNA-seq data from PDIA1-silenced MCF-7 (ERα-positive) and MDA-MB-231 (ERα-negative) breast cancer cells treated with either interferon γ (IFN-γ) or etoposide (ETO), and the obtained data were further analyzed using a variety of bioinformatic tools alongside clinical relevance assessment via Kaplan-Meier patient survival curves. The results highlighted the dual role of PDIA1 in suppressing carcinogenesis in the ERα(+) breast cancer patients by negatively regulating the response to reactive oxygen species (ROS) and promoting carcinogenesis by inducing cell cycle progression. In the ERα(−) breast cancer patients, PDIA1 prevented tumor development by modulating NF-κB and p53 activity and cell migration and induced breast cancer progression through control of cytokine signaling and the immune response. The findings reported in this study shed light on the differential pathways regulating carcinogenesis in ERα(+) and ERα(−) breast cancer patients and could help identify therapeutic targets selectively effective in ERα(+) vs. ERα(−) patients.
Collapse
Affiliation(s)
- Emyr Yosef Bakker
- School of Medicine, University of Central Lancashire, Preston, Lancashire PR1 2HE, UK
| | - Masayuki Fujii
- Department of Biological and Environmental Chemistry, Faculty of Humanity Oriented Science and Engineering, Kindai University, Iizuka, Fukuoka 820‑8555, Japan
| | | | - Constantinos Demonacos
- Faculty of Biology Medicine and Health, School of Health Science, Division of Pharmacy and Optometry, University of Manchester, Manchester M13 9PT, UK
| | - Rashed Alhammad
- Faculty of Biology Medicine and Health, School of Health Science, Division of Pharmacy and Optometry, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
8
|
Jha V, Kumari T, Manickam V, Assar Z, Olson KL, Min JK, Cho J. ERO1-PDI Redox Signaling in Health and Disease. Antioxid Redox Signal 2021; 35:1093-1115. [PMID: 34074138 PMCID: PMC8817699 DOI: 10.1089/ars.2021.0018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Significance: Protein disulfide isomerase (PDI) and endoplasmic reticulum oxidoreductase 1 (ERO1) are crucial for oxidative protein folding in the endoplasmic reticulum (ER). These enzymes are frequently overexpressed and secreted, and they contribute to the pathology of neurodegenerative, cardiovascular, and metabolic diseases. Recent Advances: Tissue-specific knockout mouse models and pharmacologic inhibitors have been developed to advance our understanding of the cell-specific functions of PDI and ERO1. In addition to their roles in protecting cells from the unfolded protein response and oxidative stress, recent studies have revealed that PDI and ERO1 also function outside of the cells. Critical Issues: Despite the well-known contributions of PDI and ERO1 to specific disease pathology, the detailed molecular and cellular mechanisms underlying these activities remain to be elucidated. Further, although PDI and ERO1 inhibitors have been identified, the results from previous studies require careful evaluation, as many of these agents are not selective and may have significant cytotoxicity. Future Directions: The functions of PDI and ERO1 in the ER have been extensively studied. Additional studies will be required to define their functions outside the ER.
Collapse
Affiliation(s)
- Vishwanath Jha
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tripti Kumari
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Vijayprakash Manickam
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Zahra Assar
- Cayman Chemical Company, Inc., Ann Arbor, Michigan, USA
| | - Kirk L Olson
- Cayman Chemical Company, Inc., Ann Arbor, Michigan, USA
| | - Jeong-Ki Min
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
9
|
Fernandes IG, de Brito CA, dos Reis VMS, Sato MN, Pereira NZ. SARS-CoV-2 and Other Respiratory Viruses: What Does Oxidative Stress Have to Do with It? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8844280. [PMID: 33381273 PMCID: PMC7757116 DOI: 10.1155/2020/8844280] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/09/2020] [Accepted: 12/13/2020] [Indexed: 02/08/2023]
Abstract
The phenomenon of oxidative stress, characterized as an imbalance in the production of reactive oxygen species and antioxidant responses, is a well-known inflammatory mechanism and constitutes an important cellular process. The relationship of viral infections, reactive species production, oxidative stress, and the antiviral response is relevant. Therefore, the aim of this review is to report studies showing how reactive oxygen species may positively or negatively affect the pathophysiology of viral infection. We focus on known respiratory viral infections, especially severe acute respiratory syndrome coronaviruses (SARS-CoVs), in an attempt to provide important information on the challenges posed by the current COVID-19 pandemic. Because antiviral therapies for severe acute respiratory syndrome coronaviruses (e.g., SARS-CoV-2) are rare, knowledge about relevant antioxidant compounds and oxidative pathways may be important for understanding viral pathogenesis and identifying possible therapeutic targets.
Collapse
Affiliation(s)
- Iara Grigoletto Fernandes
- Laboratory of Medical Investigation 56, Dermatology Department, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Cyro Alves de Brito
- Technical Division of Medical Biology, Immunology Center, Adolfo Lutz Institute, São Paulo, Brazil
| | | | - Maria Notomi Sato
- Laboratory of Medical Investigation 56, Dermatology Department, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Nátalli Zanete Pereira
- Laboratory of Medical Investigation 56, Dermatology Department, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| |
Collapse
|