1
|
Abad C, Pinal-Fernandez I, Guillou C, Bourdenet G, Drouot L, Cosette P, Giannini M, Debrut L, Jean L, Bernard S, Genty D, Zoubairi R, Remy-Jouet I, Geny B, Boitard C, Mammen A, Meyer A, Boyer O. IFNγ causes mitochondrial dysfunction and oxidative stress in myositis. Nat Commun 2024; 15:5403. [PMID: 38926363 PMCID: PMC11208592 DOI: 10.1038/s41467-024-49460-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Idiopathic inflammatory myopathies (IIMs) are severe autoimmune diseases with poorly understood pathogenesis and unmet medical needs. Here, we examine the role of interferon γ (IFNγ) using NOD female mice deficient in the inducible T cell co-stimulator (Icos), which have previously been shown to develop spontaneous IFNγ-driven myositis mimicking human disease. Using muscle proteomic and spatial transcriptomic analyses we reveal profound myofiber metabolic dysregulation in these mice. In addition, we report muscle mitochondrial abnormalities and oxidative stress in diseased mice. Supporting a pathogenic role for oxidative stress, treatment with a reactive oxygen species (ROS) buffer compound alleviated myositis, preserved muscle mitochondrial ultrastructure and respiration, and reduced inflammation. Mitochondrial anomalies and oxidative stress were diminished following anti-IFNγ treatment. Further transcriptomic analysis in IIMs patients and human myoblast in vitro studies supported the link between IFNγ and mitochondrial dysfunction observed in mice. These results suggest that mitochondrial dysfunction, ROS and inflammation are interconnected in a self-maintenance loop, opening perspectives for mitochondria therapy and/or ROS targeting drugs in myositis.
Collapse
Affiliation(s)
- Catalina Abad
- Univ Rouen Normandie, Inserm, UMR1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France
| | - Iago Pinal-Fernandez
- Muscle Disease Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Clement Guillou
- Univ Rouen Normandie, Inserm US 51, CNRS UAR 2026, HeRacLeS PISSARO, F-76000, Rouen, France
| | - Gwladys Bourdenet
- Univ Rouen Normandie, Inserm, UMR1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France
| | - Laurent Drouot
- Univ Rouen Normandie, Inserm, UMR1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France
| | - Pascal Cosette
- Univ Rouen Normandie, Inserm US 51, CNRS UAR 2026, HeRacLeS PISSARO, F-76000, Rouen, France
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, Normandie Univ, PBS UMR 6270, F-76000, Rouen, France
| | - Margherita Giannini
- Translational Medicine Federation of Strasbourg, Team 3072, Faculty of Medicine, University of Strasbourg, Strasbourg, France
- Unité exploration fonctionnelle musculaire-service de physiologie, Centre National de Référence des Maladies Auto-Immunes Systémiques Rares de l'Est et du Sud-Ouest -Service de rhumatologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Lea Debrut
- Translational Medicine Federation of Strasbourg, Team 3072, Faculty of Medicine, University of Strasbourg, Strasbourg, France
| | - Laetitia Jean
- Univ Rouen Normandie, Inserm, UMR1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France
| | - Sophie Bernard
- Univ Rouen Normandie, Inserm US51, CNRS UAR2026, HeRacLeS PRIMACEN, F-76000, Rouen, France
| | - Damien Genty
- CHU Rouen, Department of Pathology, F-76000, Rouen, France
| | - Rachid Zoubairi
- Univ Rouen Normandie, Inserm, UMR1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France
| | - Isabelle Remy-Jouet
- Univ Rouen Normandie, Inserm, UMR1096, BOSS facility, F-76000, Rouen, France
| | - Bernard Geny
- Translational Medicine Federation of Strasbourg, Team 3072, Faculty of Medicine, University of Strasbourg, Strasbourg, France
- Unité exploration fonctionnelle musculaire-service de physiologie, Centre National de Référence des Maladies Auto-Immunes Systémiques Rares de l'Est et du Sud-Ouest -Service de rhumatologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Christian Boitard
- Cochin Institute, Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - Andrew Mammen
- Muscle Disease Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alain Meyer
- Translational Medicine Federation of Strasbourg, Team 3072, Faculty of Medicine, University of Strasbourg, Strasbourg, France
- Unité exploration fonctionnelle musculaire-service de physiologie, Centre National de Référence des Maladies Auto-Immunes Systémiques Rares de l'Est et du Sud-Ouest -Service de rhumatologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Olivier Boyer
- Univ Rouen Normandie, Inserm, UMR1234, FOCIS Center of Excellence PAn'THER, F-76000, Rouen, France.
- CHU Rouen, Department of Immunology and Biotherapy, F-76000, Rouen, France.
| |
Collapse
|
2
|
Anjo SI, He Z, Hussain Z, Farooq A, McIntyre A, Laughton CA, Carvalho AN, Finelli MJ. Protein Oxidative Modifications in Neurodegenerative Diseases: From Advances in Detection and Modelling to Their Use as Disease Biomarkers. Antioxidants (Basel) 2024; 13:681. [PMID: 38929122 PMCID: PMC11200609 DOI: 10.3390/antiox13060681] [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: 05/02/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Oxidation-reduction post-translational modifications (redox-PTMs) are chemical alterations to amino acids of proteins. Redox-PTMs participate in the regulation of protein conformation, localization and function, acting as signalling effectors that impact many essential biochemical processes in the cells. Crucially, the dysregulation of redox-PTMs of proteins has been implicated in the pathophysiology of numerous human diseases, including neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. This review aims to highlight the current gaps in knowledge in the field of redox-PTMs biology and to explore new methodological advances in proteomics and computational modelling that will pave the way for a better understanding of the role and therapeutic potential of redox-PTMs of proteins in neurodegenerative diseases. Here, we summarize the main types of redox-PTMs of proteins while providing examples of their occurrence in neurodegenerative diseases and an overview of the state-of-the-art methods used for their detection. We explore the potential of novel computational modelling approaches as essential tools to obtain insights into the precise role of redox-PTMs in regulating protein structure and function. We also discuss the complex crosstalk between various PTMs that occur in living cells. Finally, we argue that redox-PTMs of proteins could be used in the future as diagnosis and prognosis biomarkers for neurodegenerative diseases.
Collapse
Affiliation(s)
- Sandra I. Anjo
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-517 Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3030-789 Coimbra, Portugal
| | - Zhicheng He
- Biodiscovery Institute, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Zohaib Hussain
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| | - Aruba Farooq
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| | - Alan McIntyre
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| | - Charles A. Laughton
- Biodiscovery Institute, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Andreia Neves Carvalho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Mattéa J. Finelli
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| |
Collapse
|
3
|
Jeong C, Kim HJ. YabJ from Staphylococcus aureus entraps chlorides within its pocket. Biochem Biophys Res Commun 2024; 710:149892. [PMID: 38581951 DOI: 10.1016/j.bbrc.2024.149892] [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: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
Chlorination is a potent disinfectant against various microorganisms, including bacteria and viruses, by inducing protein modifications and functional changes. Chlorine, in the form of sodium hypochlorite, stands out as the predominant sanitizer choice due to its cost-effectiveness and powerful antimicrobial properties. Upon exposure to chlorination, proteins undergo modifications, with amino acids experiencing alterations through the attachment of chloride or oxygen atoms. These modifications lead to shifts in protein function and the modulation of downstream signaling pathways, ultimately resulting in a bactericidal effect. However, certain survival proteins, such as chaperones or transcription factors, aid organisms in overcoming harsh chlorination conditions. The expression of YabJ, a highly conserved protein from Staphylococcus aureus, is regulated by a stress-activated sigma factor called sigma B (σB). This research revealed that S. aureus YabJ maintains its structural integrity even under intense chlorination conditions and harbors sodium hypochlorite molecules within its surface pocket. Notably, the pocket of S. aureus YabJ is primarily composed of amino acids less susceptible to chlorination-induced damage, rendering it resistant to such effects. This study elucidates how S. aureus YabJ evades the detrimental effects of chlorination and highlights its role in sequestering sodium hypochlorite within its structure. Consequently, this process enhances resilience and facilitates adaptation to challenging environmental conditions.
Collapse
Affiliation(s)
- Cheolwoo Jeong
- College of Pharmacy, Woosuk University, Wanju, 55338, Republic of Korea
| | - Hyo Jung Kim
- College of Pharmacy, Woosuk University, Wanju, 55338, Republic of Korea.
| |
Collapse
|
4
|
Rio P, Gasbarrini A, Gambassi G, Cianci R. Pollutants, microbiota and immune system: frenemies within the gut. Front Public Health 2024; 12:1285186. [PMID: 38799688 PMCID: PMC11116734 DOI: 10.3389/fpubh.2024.1285186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 04/24/2024] [Indexed: 05/29/2024] Open
Abstract
Pollution is a critical concern of modern society for its heterogeneous effects on human health, despite a widespread lack of awareness. Environmental pollutants promote several pathologies through different molecular mechanisms. Pollutants can affect the immune system and related pathways, perturbing its regulation and triggering pro-inflammatory responses. The exposure to several pollutants also leads to alterations in gut microbiota with a decreasing abundance of beneficial microbes, such as short-chain fatty acid-producing bacteria, and an overgrowth of pro-inflammatory species. The subsequent intestinal barrier dysfunction, together with oxidative stress and increased inflammatory responses, plays a role in the pathogenesis of gastrointestinal inflammatory diseases. Moreover, pollutants encourage the inflammation-dysplasia-carcinoma sequence through various mechanisms, such as oxidative stress, dysregulation of cellular signalling pathways, cell cycle impairment and genomic instability. In this narrative review, we will describe the interplay between pollutants, gut microbiota, and the immune system, focusing on their relationship with inflammatory bowel diseases and colorectal cancer. Understanding the biological mechanisms underlying the health-to-disease transition may allow the design of public health policies aimed at reducing the burden of disease related to pollutants.
Collapse
Affiliation(s)
| | | | | | - Rossella Cianci
- Department of Translational Medicine and Surgery, Catholic University of Sacred Heart, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| |
Collapse
|
5
|
Khanam A, Alouffi S, Alyahyawi AR, Husain A, Khan S, Alharazi T, Akasha R, Khan H, Shahab U, Ahmad S. Generation of autoantibodies against glycated fibrinogen: Role in diabetic nephropathy and retinopathy. Anal Biochem 2024; 685:115393. [PMID: 37977213 DOI: 10.1016/j.ab.2023.115393] [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: 08/30/2023] [Revised: 11/01/2023] [Accepted: 11/11/2023] [Indexed: 11/19/2023]
Abstract
The process of glycation, characterized by the non-enzymatic reaction between sugars and free amino groups on biomolecules, is a key contributor to the development and progression of both microvascular and macrovascular complications associated with diabetes, particularly due to persistent hyperglycemia. This glycation process gives rise to advanced glycation end products (AGEs), which play a central role in the pathophysiology of diabetes complications, including nephropathy. The d-ribose-mediated glycation of fibrinogen plays a central role in the pathogenesis of diabetes nephropathy (DN) and retinopathy (DR) by the generation and accumulation of advanced glycation end products (AGEs). Glycated fibrinogen with d-ribose (Rb-gly-Fb) induces structural changes that trigger an autoimmune response by generating and exposing neoepitopes on fibrinogen molecules. The present research is designed to investigate the prevalence of autoantibodies against Rb-gly-Fb in individuals with type 2 diabetes mellitus (T2DM), DN & DR. Direct binding ELISA was used to test the binding affinity of autoantibodies from patients' sera against Rb-gly-Fb and competitive ELISA was used to confirm the direct binding findings by checking the bindings of isolated IgG against Rb-gly-Fb and its native conformer. In comparison to healthy subjects, 32% of T2DM, 67% of DN and 57.85% of DR patients' samples demonstrated a strong binding affinity towards Rb-gly-Fb. Both native and Rb-gly-Fb binding by healthy subjects (HS) sera were non-significant (p > 0.05). Furthermore, the early, intermediate, and end products of glycation have been assessed through biochemical and physicochemical analysis. The biochemical markers in the patient groups were also significant (p < 0.05) in comparison to the HS group. This study not only establishes the prevalence of autoantibodies against d-ribose glycated fibrinogen in DN but also highlights the potential of glycated fibrinogen as a biomarker for the detection of DN and/or DR. These insights may open new avenues for research into novel therapeutic strategies and the prevention of diabetes-related nephropathy and retinopathy.
Collapse
Affiliation(s)
- Afreen Khanam
- Department of Biosciences, Faculty of Sciences, Integral University, Lucknow, 226026, India; Department of Biotechnology & Life Sciences, Institute of Biomedical Education & Research, Mangalayatan University, Aligarh, 202146, India
| | - Sultan Alouffi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail- 2440, Saudi Arabia
| | - Amjad R Alyahyawi
- Department of Diagnostic Radiology, College of Applied Medical Science, University of Hail, Ha'il, 2440, Saudi Arabia; Centre for Nuclear and Radiation Physics, Department of Physics, University of Surrey, Guildford, GU2 7XH, United Kingdom
| | - Arbab Husain
- Department of Biosciences, Faculty of Sciences, Integral University, Lucknow, 226026, India; Department of Biotechnology & Life Sciences, Institute of Biomedical Education & Research, Mangalayatan University, Aligarh, 202146, India
| | - Saif Khan
- Department of Basic Dental and Medical Sciences, College of Dentistry, University of Hail, Saudi Arabia
| | - Talal Alharazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail- 2440, Saudi Arabia
| | - Rihab Akasha
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail- 2440, Saudi Arabia
| | - Hamda Khan
- Department of Biochemistry, Jawahar Lal Nehru Medical College, Aligarh Muslim University, Aligarh, 202002, India
| | - Uzma Shahab
- Department of Biochemistry, King George Medical University, Lucknow, 226003, India
| | - Saheem Ahmad
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail- 2440, Saudi Arabia.
| |
Collapse
|
6
|
Senturk A, Alver A, Karkucak M, Küçük M, Ahmadi Rendi T. Oxidative modification of carbonic anhydrase by peroxynitrite trigger immune response in mice and rheumatic disease patients. Am J Med Sci 2023; 366:438-448. [PMID: 37678670 DOI: 10.1016/j.amjms.2023.09.002] [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: 02/01/2023] [Revised: 08/11/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Carbonic anhydrases (CA) are metalloenzymes with wide tissue distribution, involved in many important physiological processes, and in some rheumatic diseases, autoantibodies are formed against these enzymes. Recent studies have suggested that oxidative stress triggers anti-CA antibody formation. In this study, we aimed to investigate the effects of modification with oxidative/nitrosative stress end products on CA antigenicity in mice and the relationship between the modified CA autoantibodies and oxidant-antioxidant status in patients with rheumatoid arthritis (RA) and Sjögren's syndrome (SjS). METHODS CA I and CA II isoenzymes were isolated from human erythrocytes and modified with 4-hydroxynonenal (4-HNE), malondialdehyde (MDA), and peroxynitrite (PN). Balb-c mice were immunized with these agents to determine the effects of modification on CA antigenicity. The autoantibody titers of modified CA isoenzymes were detected in patients. In addition MDA, 4-HNE, 3-nitrotyrosine (3-NT), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) activities were measured to assess the oxidant-antioxidant status in patients. RESULTS Modifications of carbonic anhydrase with oxidative stress end products, HNE, MDA and PN, lead to alterations in the immune response to these enzymes in mice. It was found that HNE and MDA decreased the antigenicity while PN increased. In addition, PN-modified CA autoantibody levels were found to be significantly different in both RA and SjS patients compared to their controls (p<0.05). CONCLUSIONS PN modifications can also trigger an immune response against CA isoenzymes in mice, and PN-modified CA I and CA II autoantibody titers were found at a significantly high level in both RA and SjS patients.
Collapse
Affiliation(s)
- Ayse Senturk
- Macka Vocational School, Karadeniz Technical University, Trabzon 61750, Türkiye.
| | - Ahmet Alver
- Faculty of Medicine, Department of Medical Biochemistry, Karadeniz Technical University, Trabzon, Türkiye
| | - Murat Karkucak
- Faculty of Medicine, Department of Physical Medicine and Rehabilitation, Karadeniz Technical University, Trabzon, Türkiye
| | - Murat Küçük
- Faculty of Science, Department of Chemistry, Karadeniz Technical University, Trabzon, Türkiye
| | - Taghi Ahmadi Rendi
- Graduate School of Medical Science, Department of Medical Biochemistry, Karadeniz Technical University, Trabzon, Türkiye; Faculty of Medicine, Department of Medical Biochemistry, Istanbul University, Istanbul, Turkey
| |
Collapse
|
7
|
Riitano G, Recalchi S, Capozzi A, Manganelli V, Misasi R, Garofalo T, Sorice M, Longo A. The Role of Autophagy as a Trigger of Post-Translational Modifications of Proteins and Extracellular Vesicles in the Pathogenesis of Rheumatoid Arthritis. Int J Mol Sci 2023; 24:12764. [PMID: 37628944 PMCID: PMC10454292 DOI: 10.3390/ijms241612764] [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: 07/12/2023] [Revised: 08/11/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease, characterized by persistent joint inflammation, leading to cartilage and bone destruction. Autoantibody production is directed to post-translational modified (PTM) proteins, i.e., citrullinated or carbamylated. Autophagy may be the common feature in several types of stress (smoking, joint injury, and infections) and may be involved in post-translational modifications (PTMs) in proteins and the generation of citrullinated and carbamylated peptides recognized by the immune system in RA patients, with a consequent breakage of tolerance. Interestingly, autophagy actively provides information to neighboring cells via a process called secretory autophagy. Secretory autophagy combines the autophagy machinery with the secretion of cellular content via extracellular vesicles (EVs). A role for exosomes in RA pathogenesis has been recently demonstrated. Exosomes are involved in intercellular communications, and upregulated proteins and RNAs may contribute to the development of inflammatory arthritis and the progression of RA. In RA, most of the exosomes are produced by leukocytes and synoviocytes, which are loaded with PTM proteins, mainly citrullinated proteins, inflammatory molecules, and enzymes that are implicated in RA pathogenesis. Microvesicles derived from cell plasma membrane may also be loaded with PTM proteins, playing a role in the immunopathogenesis of RA. An analysis of changes in EV profiles, including PTM proteins, could be a useful tool for the prevention of inflammation in RA patients and help in the discovery of personalized medicine.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Maurizio Sorice
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy; (G.R.); (S.R.); (A.C.); (V.M.); (R.M.); (T.G.); (A.L.)
| | | |
Collapse
|
8
|
Sorice M, Profumo E, Capozzi A, Recalchi S, Riitano G, Di Veroli B, Saso L, Buttari B. Oxidative Stress as a Regulatory Checkpoint in the Production of Antiphospholipid Autoantibodies: The Protective Role of NRF2 Pathway. Biomolecules 2023; 13:1221. [PMID: 37627286 PMCID: PMC10452087 DOI: 10.3390/biom13081221] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/13/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Oxidative stress is a well-known hallmark of Antiphospholipid Antibody Syndrome (APS), a systemic autoimmune disease characterized by arterial and venous thrombosis and/or pregnancy morbidity. Oxidative stress may affect various signaling pathways and biological processes, promoting dysfunctional immune responses and inflammation, inducing apoptosis, deregulating autophagy and impairing mitochondrial function. The chronic oxidative stress and the dysregulation of the immune system leads to the loss of tolerance, which drives autoantibody production and inflammation with the development of endothelial dysfunction. In particular, anti-phospholipid antibodies (aPL), which target phospholipids and/or phospholipid binding proteins, mainly β-glycoprotein I (β-GPI), play a functional role in the cell signal transduction pathway(s), thus contributing to oxidative stress and thrombotic events. An oxidation-antioxidant imbalance may be detected in the blood of patients with APS as a reflection of disease progression. This review focuses on functional evidence highlighting the role of oxidative stress in the initiation and progression of APS. The protective role of food supplements and Nuclear Factor Erythroid 2-Related Factor 2 (NRF2) activators in APS patients will be summarized to point out the potential of these therapeutic approaches to reduce APS-related clinical complications.
Collapse
Affiliation(s)
- Maurizio Sorice
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (M.S.); (A.C.); (S.R.); (G.R.)
| | - Elisabetta Profumo
- Department of Cardiovascular and Endocrine-metabolic Diseases and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy; (E.P.); (B.D.V.)
| | - Antonella Capozzi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (M.S.); (A.C.); (S.R.); (G.R.)
| | - Serena Recalchi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (M.S.); (A.C.); (S.R.); (G.R.)
| | - Gloria Riitano
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (M.S.); (A.C.); (S.R.); (G.R.)
| | - Benedetta Di Veroli
- Department of Cardiovascular and Endocrine-metabolic Diseases and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy; (E.P.); (B.D.V.)
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy;
| | - Brigitta Buttari
- Department of Cardiovascular and Endocrine-metabolic Diseases and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy; (E.P.); (B.D.V.)
| |
Collapse
|
9
|
Batsalova T, Dzhambazov B. Significance of Type II Collagen Posttranslational Modifications: From Autoantigenesis to Improved Diagnosis and Treatment of Rheumatoid Arthritis. Int J Mol Sci 2023; 24:9884. [PMID: 37373030 PMCID: PMC10298457 DOI: 10.3390/ijms24129884] [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: 05/19/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Collagen type II (COL2), the main structural protein of hyaline cartilage, is considerably affected by autoimmune responses associated with the pathogenesis of rheumatoid arthritis (RA). Posttranslational modifications (PTMs) play a significant role in the formation of the COL2 molecule and supramolecular fibril organization, and thus, support COL2 function, which is crucial for normal cartilage structure and physiology. Conversely, the specific PTMs of the protein (carbamylation, glycosylation, citrullination, oxidative modifications and others) have been implicated in RA autoimmunity. The discovery of the anti-citrullinated protein response in RA, which includes anti-citrullinated COL2 reactivity, has led to the development of improved diagnostic assays and classification criteria for the disease. The induction of immunological tolerance using modified COL2 peptides has been highlighted as a potentially effective strategy for RA therapy. Therefore, the aim of this review is to summarize the recent knowledge on COL2 posttranslational modifications with relevance to RA pathophysiology, diagnosis and treatment. The significance of COL2 PTMs as a source of neo-antigens that activate immunity leading to or sustaining RA autoimmunity is discussed.
Collapse
Affiliation(s)
| | - Balik Dzhambazov
- Faculty of Biology, Paisii Hilendarski University of Plovdiv, 24 Tsar Assen Str., 4000 Plovdiv, Bulgaria;
| |
Collapse
|
10
|
Esposito I, Kontra I, Giacomassi C, Manou-Stathopoulou S, Brown J, Stratton R, Verykokou G, Buccafusca R, Stevens M, Nissim A, Lewis MJ, Pfeffer PE. Identification of autoantigens and their potential post-translational modification in EGPA and severe eosinophilic asthma. Front Immunol 2023; 14:1164941. [PMID: 37334358 PMCID: PMC10272393 DOI: 10.3389/fimmu.2023.1164941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/18/2023] [Indexed: 06/20/2023] Open
Abstract
Background The chronic airway inflammation in severe eosinophilic asthma (SEA) suggests potential autoimmune aetiology with unidentified autoantibodies analogous to myeloperoxidase (MPO) in ANCA-positive EGPA (eosinophilic granulomatosis with polyangiitis). Previous research has shown that oxidative post-translational modification (oxPTM) of proteins is an important mechanism by which autoantibody responses may escape immune tolerance. Autoantibodies to oxPTM autoantigens in SEA have not previously been studied. Methods Patients with EGPA and SEA were recruited as well as healthy control participants. Autoantigen agnostic approach: Participant serum was incubated with slides of unstimulated and PMA-stimulated neutrophils and eosinophils, and autoantibodies to granulocytes were identified by immunofluorescence with anti-human IgG FITC antibody. Target autoantigen approach: Candidate proteins were identified from previous literature and FANTOM5 gene set analysis for eosinophil expressed proteins. Serum IgG autoantibodies to these proteins, in native and oxPTM form, were detected by indirect ELISA. Results Immunofluorescence studies showed that serum from patients with known ANCA stained for IgG against neutrophils as expected. In addition, serum from 9 of 17 tested SEA patients stained for IgG to PMA-stimulated neutrophils undergoing NETosis. Immunofluorescent staining of eosinophil slides was evident with serum from all participants (healthy and with eosinophilic disease) with diffuse cytoplasmic staining except for one SEA individual in whom subtle nuclear staining was evident. FANTOM5 gene set analysis identified TREM1 (triggering receptor expressed on myeloid cells 1) and IL-1 receptor 2 (IL1R2) as eosinophil-specific targets to test for autoantibody responses in addition to MPO, eosinophil peroxidase (EPX), and Collagen-V identified from previous literature. Indirect ELISAs found high concentrations of serum autoantibodies to Collagen-V, MPO, and TREM1 in a higher proportion of SEA patients than healthy controls. High concentrations of serum autoantibodies to EPX were evident in serum from both healthy and SEA participants. The proportion of patients with positive autoantibody ELISAs was not increased when examining oxPTM compared to native proteins. Discussion Although none of the target proteins studied showed high sensitivity for SEA, the high proportion of patients positive for at least one serum autoantibody shows the potential of more research on autoantibody serology to improve diagnostic testing for severe asthma. Clinical trial registration ClinicalTrials.gov, identifier, NCT04671446.
Collapse
Affiliation(s)
- Ilaria Esposito
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Ioanna Kontra
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Chiara Giacomassi
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- Department of Rheumatology, Royal Free NHS Foundation Trust, London, United Kingdom
| | | | - James Brown
- Department of Respiratory Medicine, Royal Free NHS Foundation Trust, London, United Kingdom
| | - Richard Stratton
- Department of Rheumatology, Royal Free NHS Foundation Trust, London, United Kingdom
- Centre for Rheumatology, University College London, London, United Kingdom
| | - Galateia Verykokou
- Department of Respiratory Medicine, Barts Health NHS Trust, London, United Kingdom
| | - Roberto Buccafusca
- School of Physical and Chemical Sciences, Queen Mary University of London, London, United Kingdom
| | - Michael Stevens
- Department of Clinical Immunology, Barts Health NHS Trust, London, United Kingdom
- Department of Clinical Immunology, University Hospitals Sussex NHS Foundation Trust, Brighton, United Kingdom
| | - Ahuva Nissim
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Myles J. Lewis
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Paul E. Pfeffer
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- Department of Respiratory Medicine, Barts Health NHS Trust, London, United Kingdom
| |
Collapse
|
11
|
Bardelčíková A, Šoltys J, Mojžiš J. Oxidative Stress, Inflammation and Colorectal Cancer: An Overview. Antioxidants (Basel) 2023; 12:antiox12040901. [PMID: 37107276 PMCID: PMC10135609 DOI: 10.3390/antiox12040901] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Colorectal cancer (CRC) represents the second leading cause of cancer-related deaths worldwide. The pathogenesis of CRC is a complex multistep process. Among other factors, inflammation and oxidative stress (OS) have been reported to be involved in the initiation and development of CRC. Although OS plays a vital part in the life of all organisms, its long-term effects on the human body may be involved in the development of different chronic diseases, including cancer diseases. Chronic OS can lead to the oxidation of biomolecules (nucleic acids, lipids and proteins) or the activation of inflammatory signaling pathways, resulting in the activation of several transcription factors or the dysregulation of gene and protein expression followed by tumor initiation or cancer cell survival. In addition, it is well known that chronic intestinal diseases such as inflammatory bowel disease (IBD) are associated with an increased risk of cancer, and a link between OS and IBD initiation and progression has been reported. This review focuses on the role of oxidative stress as a causative agent of inflammation in colorectal cancer.
Collapse
Affiliation(s)
- Annamária Bardelčíková
- Department of Pharmacology, Medical Faculty of University of Pavol Jozef Šafárik in Košice, Tr. SNP 1, 040 11 Košice, Slovakia
| | - Jindřich Šoltys
- Institute of Parasitology, Slovak Academy of Science, Hlinkova 3, 040 01 Košice, Slovakia
| | - Ján Mojžiš
- Department of Pharmacology, Medical Faculty of University of Pavol Jozef Šafárik in Košice, Tr. SNP 1, 040 11 Košice, Slovakia
| |
Collapse
|
12
|
Clemen R, Arlt K, von Woedtke T, Bekeschus S. Gas Plasma Protein Oxidation Increases Immunogenicity and Human Antigen-Presenting Cell Maturation and Activation. Vaccines (Basel) 2022; 10:1814. [PMID: 36366323 PMCID: PMC9698879 DOI: 10.3390/vaccines10111814] [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: 09/12/2022] [Revised: 10/24/2022] [Accepted: 10/24/2022] [Indexed: 08/06/2023] Open
Abstract
Protein vaccines rely on eliciting immune responses. Inflammation is a prerequisite for immune responses to control infection and cancer but is also associated with disease onset. Reactive oxygen species (ROSs) are central during inflammation and are capable of inducing non-enzymatic oxidative protein modifications (oxMods) associated with chronic disease, which alter the functionality or immunogenicity of proteins that are relevant in cancer immunotherapy. Specifically, antigen-presenting cells (APCs) take up and degrade extracellular native and oxidized proteins to induce adaptive immune responses. However, it is less clear how oxMods alter the protein's immunogenicity, especially in inflammation-related short-lived reactive species. Gas plasma technology simultaneously generates a multitude of ROSs to modify protein antigens in a targeted and controlled manner to study the immunogenicity of oxMods. As model proteins relevant to chronic inflammation and cancer, we used gas plasma-treated insulin and CXCL8. We added those native or oxidized proteins to human THP-1 monocytes or primary monocyte-derived cells (moDCs). Both oxidized proteins caused concentration-independent maturation phenotype alterations in moDCs and THP-1 cells concerning surface marker expression and chemokine and cytokine secretion profiles. Interestingly, concentration-matched H2O2-treated proteins did not recapitulate the effects of gas plasma, suggesting sufficiently short diffusion distances for the short-lived reactive species to modify proteins. Our data provide evidence of dendric cell maturation and activation upon exposure to gas plasma- but not H2O2-modified model proteins. The biological consequences of these findings need to be elucidated in future inflammation and cancer disease models.
Collapse
|
13
|
Do Circulating Redox Biomarkers Have Diagnostic Significance in Alcohol-Intoxicated People? Int J Mol Sci 2022; 23:ijms231911808. [PMID: 36233115 PMCID: PMC9569923 DOI: 10.3390/ijms231911808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/28/2022] [Accepted: 10/01/2022] [Indexed: 11/24/2022] Open
Abstract
The toxic properties of ethanol are inextricably linked to oxidative stress. Despite many reports on the effects of alcohol dependence on blood redox homeostasis, there are no data on the oxidative stress profile in alcohol-poisoned cases. There are also no data on the diagnostic usefulness of redox biomarkers determined post-mortem in various biological fluids. This work investigates the utility of enzymatic and non-enzymatic antioxidant barrier, redox status, and oxidative/nitrosative stress biomarkers in different biological fluids (such as blood, urine, vitreous humor, and cerebrospinal fluid) in the post-mortem study of patients with acute alcohol intoxication. The study group included those who died due to acute ethanol intoxication (n = 22). The research showed a significant increase in glutathione peroxidase activity, total antioxidant status, ferric reducing antioxidant power, and tryptophan concentration only in the study group’s urine compared to the control. In other circulating fluids, both antioxidant enzyme activities and glycoxidation product concentrations were not significantly different in individuals who died of alcohol overdose compared with those who died suddenly. We also did not observe a connection between oxidation–reduction balance and the amount of alcohol consumed before death. These unexpected observations may be caused by irreversible post-mortem changes occurring at the cellular level due to autolysis and putrefaction. In summary, the use of circulating body fluids to assess redox homeostasis is limited in the post-mortem analysis. Our results indicate the increased stability of urine collected post mortem compared to other circulating bioliquids. Further studies are needed to assess the intensity of oxidative and carbonyl stress in ethanol-damaged organs and the effects of post-mortem processes on cellular redox balance.
Collapse
|
14
|
Ferrara F, Cordone V, Pecorelli A, Benedusi M, Pambianchi E, Guiotto A, Vallese A, Cervellati F, Valacchi G. Ubiquitination as a key regulatory mechanism for O 3-induced cutaneous redox inflammasome activation. Redox Biol 2022; 56:102440. [PMID: 36027676 PMCID: PMC9425076 DOI: 10.1016/j.redox.2022.102440] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 10/26/2022] Open
Abstract
NLRP1 is one of the major inflammasomes modulating the cutaneous inflammatory responses and therefore linked to a variety of cutaneous conditions. Although NLRP1 has been the first inflammasome to be discovered, only in the past years a significant progress was achieved in understanding the molecular mechanism and the stimuli behind its activation. In the past decades a crescent number of studies have highlighted the role of air pollutants as Particulate Matter (PM), Cigarette Smoke (CS) and Ozone (O3) as trigger stimuli for inflammasomes activation, especially via Reactive Oxygen Species (ROS) mediators. However, whether NLRP1 can be modulated by air pollutants via oxidative stress and the mechanism behind its activation is still poorly understood. Here we report for the first time that O3, one of the most toxic pollutants, activates the NLRP1 inflammasome in human keratinocytes via oxidative stress mediators as hydrogen peroxide (H2O2) and 4-hydroxy-nonenal (4HNE). Our data suggest that NLRP1 represents a target protein for 4HNE adduction that possibly leads to its proteasomal degradation and activation via the possible involvement of E3 ubiquitin ligase UBR2. Of note, Catalase (Cat) treatment prevented inflammasome assemble and inflammatory cytokines release as well as NLRP1 ubiquitination in human keratinocytes upon O3 exposure. The present work is a mechanistic study that follows our previous work where we have showed the ability of O3 to induce cutaneous inflammasome activation in humans exposed to this pollutant. In conclusion, our results suggest that O3 triggers the cutaneous NLRP1 inflammasome activation by ubiquitination and redox mechanism.
Collapse
Affiliation(s)
- Francesca Ferrara
- Dept. of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Valeria Cordone
- Dept. of Environmental Sciences and Prevention, University of Ferrara, Ferrara, Italy
| | - Alessandra Pecorelli
- Plants for Human Health Institute, Animal Sciences Dept., NC Research Campus, NC State University, Kannapolis, NC, USA
| | - Mascia Benedusi
- Dept. of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Erika Pambianchi
- Plants for Human Health Institute, Animal Sciences Dept., NC Research Campus, NC State University, Kannapolis, NC, USA.
| | - Anna Guiotto
- Dept. of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy; Plants for Human Health Institute, Animal Sciences Dept., NC Research Campus, NC State University, Kannapolis, NC, USA
| | - Andrea Vallese
- Dept. of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Franco Cervellati
- Dept. of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Giuseppe Valacchi
- Plants for Human Health Institute, Animal Sciences Dept., NC Research Campus, NC State University, Kannapolis, NC, USA; Dept. of Environmental Sciences and Prevention, University of Ferrara, Ferrara, Italy; Dept. of Food and Nutrition, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Gostomska-Pampuch K, Gamian A, Rawicz-Pruszyński K, Gęca K, Tkaczuk-Włach J, Jonik I, Ożga K, Staniszewska M. Proteins in human body fluids contain in vivo antigen analog of the melibiose-derived glycation product: MAGE. Sci Rep 2022; 12:7520. [PMID: 35525899 PMCID: PMC9079080 DOI: 10.1038/s41598-022-11638-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/27/2022] [Indexed: 01/16/2023] Open
Abstract
Melibiose-derived AGE (MAGE) is an advanced glycation end-product formed in vitro in anhydrous conditions on proteins and protein-free amino acids during glycation with melibiose. Our previous studies revealed the presence of MAGE antigen in the human body and tissues of several other species, including muscles, fat, extracellular matrix, and blood. MAGE is also antigenic and induces generation of anti-MAGE antibody. The aim of this paper was to identify the proteins modified by MAGE present in human body fluids, such as serum, plasma, and peritoneal fluids. The protein-bound MAGE formed in vivo has been isolated from human blood using affinity chromatography on the resin with an immobilized anti-MAGE monoclonal antibody. Using mass spectrometry and immunochemistry it has been established that MAGE epitope is present on several human blood proteins including serum albumin, IgG, and IgA. In serum of diabetic patients, mainly the albumin and IgG were modified by MAGE, while in healthy subjects IgG and IgA carried this modification, suggesting the novel AGE can impact protein structure, contribute to auto-immunogenicity, and affect function of immunoglobulins. Some proteins in peritoneal fluid from cancer patients modified with MAGE were also observed and it indicates a potential role of MAGE in cancer.
Collapse
Affiliation(s)
- Kinga Gostomska-Pampuch
- Department of Biochemistry and Immunochemistry, Wroclaw Medical University, Chalubinskiego 10, 50-368, Wrocław, Poland
- Laboratory of Medical Microbiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wrocław, Poland
| | - Andrzej Gamian
- Laboratory of Medical Microbiology, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wrocław, Poland
| | - Karol Rawicz-Pruszyński
- Department of Surgical Oncology, Medical University of Lublin, Radziwillowska 13, 20-080, Lublin, Poland
| | - Katarzyna Gęca
- Department of Surgical Oncology, Medical University of Lublin, Radziwillowska 13, 20-080, Lublin, Poland
| | - Joanna Tkaczuk-Włach
- Diagnostic Techniques Unit, Collegium Maximum, Medical University of Lublin, Staszica 4/6, 20-081, Lublin, Poland
| | - Ilona Jonik
- Faculty of Science and Health, The John Paul II Catholic University of Lublin, Konstantynów 1J, 20-708, Lublin, Poland
| | - Kinga Ożga
- Faculty of Science and Health, The John Paul II Catholic University of Lublin, Konstantynów 1J, 20-708, Lublin, Poland
| | - Magdalena Staniszewska
- Faculty of Science and Health, The John Paul II Catholic University of Lublin, Konstantynów 1J, 20-708, Lublin, Poland.
| |
Collapse
|
16
|
Post-Translational Modifications of ATG4B in the Regulation of Autophagy. Cells 2022; 11:cells11081330. [PMID: 35456009 PMCID: PMC9025542 DOI: 10.3390/cells11081330] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/16/2022] Open
Abstract
Autophagy plays a key role in eliminating and recycling cellular components in response to stress, including starvation. Dysregulation of autophagy is observed in various diseases, including neurodegenerative diseases, cancer, and diabetes. Autophagy is tightly regulated by autophagy-related (ATG) proteins. Autophagy-related 4 (ATG4) is the sole cysteine protease, and four homologs (ATG4A–D) have been identified in mammals. These proteins have two domains: catalytic and short fingers. ATG4 facilitates autophagy by promoting autophagosome maturation through reversible lipidation and delipidation of seven autophagy-related 8 (ATG8) homologs, including microtubule-associated protein 1-light chain 3 (LC3) and GABA type A receptor-associated protein (GABARAP). Each ATG4 homolog shows a preference for a specific ATG8 homolog. Post-translational modifications of ATG4, including phosphorylation/dephosphorylation, O-GlcNAcylation, oxidation, S-nitrosylation, ubiquitination, and proteolytic cleavage, regulate its activity and ATG8 processing, thus modulating its autophagic activity. We reviewed recent advances in our understanding of the effect of post-translational modification on the regulation, activity, and function of ATG4, the main protease that controls autophagy.
Collapse
|
17
|
Basistyi VS, Frederich JH. Pyridazine N-Oxides as Photoactivatable Surrogates for Reactive Oxygen Species. Org Lett 2022; 24:1907-1912. [PMID: 35262355 PMCID: PMC10559729 DOI: 10.1021/acs.orglett.2c00227] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A method for the photoinduced evolution of atomic oxygen from pyridazine N-oxides was developed. This underexplored oxygen allotrope mediates arene C-H oxidation within complex, polyfunctional molecules. A water-soluble pyridazine N-oxide was also developed and shown to promote photoinduced DNA cleavage in aqueous solution. Taken together, these studies highlight the utility of pyridazine N-oxides as photoactivatable O(3P) precursors for applications in organic synthesis and chemical biology.
Collapse
Affiliation(s)
- Vitalii S Basistyi
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| | - James H Frederich
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| |
Collapse
|
18
|
Marrugo Padilla A, Rizzo G, Smaldini PL, Vaccaro J, Méndez Cuadro D, Rodríguez Cavallo E, Docena GH. Carbonylation induced by antibiotic and pesticide residues on casein increases its IgE binding and allergenicity. Free Radic Res 2022; 56:28-39. [DOI: 10.1080/10715762.2022.2032020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Albeiro Marrugo Padilla
- Analytical Chemistry and Biomedicine Group. Campus of Zaragocilla ancient building Cread. University of Cartagena. Cartagena-Colombia.
| | - Gastón Rizzo
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado a CIC PBA, Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, La Plata, Argentina
| | - Paola L. Smaldini
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado a CIC PBA, Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, La Plata, Argentina
| | - Julián Vaccaro
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado a CIC PBA, Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, La Plata, Argentina
| | - Darío Méndez Cuadro
- Analytical Chemistry and Biomedicine Group. Campus of Zaragocilla ancient building Cread. University of Cartagena. Cartagena-Colombia.
| | - Erika Rodríguez Cavallo
- Analytical Chemistry and Biomedicine Group. Campus of Zaragocilla ancient building Cread. University of Cartagena. Cartagena-Colombia.
| | - Guillermo H. Docena
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado a CIC PBA, Facultad de Ciencias Exactas, Departamento de Ciencias Biológicas, La Plata, Argentina
| |
Collapse
|
19
|
The Immunogenetics of Morphea and Lichen Sclerosus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1367:155-172. [DOI: 10.1007/978-3-030-92616-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
20
|
Vakilian M. A review on the effect of prolyl isomerization on immune response aberration and hypersensitivity reactions: A unifying hypothesis. Clin Immunol 2021; 234:108896. [PMID: 34848356 DOI: 10.1016/j.clim.2021.108896] [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: 08/27/2021] [Revised: 11/19/2021] [Accepted: 11/21/2021] [Indexed: 12/01/2022]
Abstract
Little is known about the causes and mechanisms of ectopic immune responses, including different types of hypersensitivity, superantigens, and cytokine storms. Two of the most questionable phenomena observed in immunology are why the intensity and extent of immune responses to different antigens are different, and why some self-antigens are attacked as foreign. The secondary structure of the peptides involved in the immune system, such as the epitope-paratope interfaces plays a pivotal role in the resulting immune responses. Prolyl cis/trans isomerization plays a fundamental role in the form of the secondary structure and the folding of proteins. This review covers some of the emerging evidence indicating the impact of prolyl isomerization on protein conformation, aberration of immune responses, and the development of hypersensitivity reactions.
Collapse
Affiliation(s)
- Mehrdad Vakilian
- Department of Cell Biology, Genetics and Physiology, University of Malaga (UMA), The Institute of Biomedical Research in Malaga (IBIMA), Málaga, Spain.
| |
Collapse
|
21
|
Jennings EQ, Fritz KS, Galligan JJ. Biochemical genesis of enzymatic and non-enzymatic post-translational modifications. Mol Aspects Med 2021; 86:101053. [PMID: 34838336 PMCID: PMC9126990 DOI: 10.1016/j.mam.2021.101053] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/07/2021] [Accepted: 11/16/2021] [Indexed: 12/20/2022]
Abstract
Post-translational modifications (PTMs) alter protein structure, function, and localization and play a pivotal role in physiological and pathophysiological conditions. Many PTMs arise from endogenous metabolic intermediates and serve as sensors for metabolic feedback to maintain cell growth and homeostasis. A key feature to PTMs is their biochemical genesis, which can result from either non-enzymatic adduction (nPTMs) or through enzyme-catalyzed reactions (ePTMs). The abundance and site-specificity of PTMs are determined by dedicated classes of enzymes that add (writers) or remove (erasers) the chemical addition. In this review we will highlight the biochemical genesis and regulation of a few of the 700+ PTMs that have been identified.
Collapse
Affiliation(s)
- Erin Q Jennings
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Kristofer S Fritz
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - James J Galligan
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
22
|
Ravi B, Kanwar P, Sanyal SK, Bheri M, Pandey GK. VDACs: An Outlook on Biochemical Regulation and Function in Animal and Plant Systems. Front Physiol 2021; 12:683920. [PMID: 34421635 PMCID: PMC8375762 DOI: 10.3389/fphys.2021.683920] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
The voltage-dependent anion channels (VDACs) are the most abundant proteins present on the outer mitochondrial membrane. They serve a myriad of functions ranging from energy and metabolite exchange to highly debatable roles in apoptosis. Their role in molecular transport puts them on the center stage as communicators between cytoplasmic and mitochondrial signaling events. Beyond their general role as interchangeable pores, members of this family may exhibit specific functions. Even after nearly five decades of their discovery, their role in plant systems is still a new and rapidly emerging field. The information on biochemical regulation of VDACs is limited. Various interacting proteins and post-translational modifications (PTMs) modulate VDAC functions, amongst these, phosphorylation is quite noticeable. In this review, we have tried to give a glimpse of the recent advancements in the biochemical/interactional regulation of plant VDACs. We also cover a critical analysis on the importance of PTMs in the functional regulation of VDACs. Besides, the review also encompasses numerous studies which can identify VDACs as a connecting link between Ca2+ and reactive oxygen species signaling in special reference to the plant systems.
Collapse
Affiliation(s)
| | | | | | | | - Girdhar K. Pandey
- Department of Plant Molecular Biology, University of Delhi, New Delhi, India
| |
Collapse
|
23
|
Talha M, Mir AR, Habib S, Abidi M, Warsi MS, Islam S. Hydroxyl radical induced structural perturbations make insulin highly immunogenic and generate an auto-immune response in type 2 diabetes mellitus. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 255:119640. [PMID: 33744841 DOI: 10.1016/j.saa.2021.119640] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/18/2021] [Accepted: 02/21/2021] [Indexed: 06/12/2023]
Abstract
Reactive oxygen species (ROS) cause oxidative damage to proteins and generate deleterious by-products which induce a breakdown of immune tolerance and produce antibodies against host macromolecules with implication in human diseases. This study characterizes the hydroxyl radical (OH) modifications of insulin, evaluates its cytotoxicity and immunogenicity, and probes its role in type 2 diabetes (T2DM) autoimmunity. The results demonstrate susceptibility of insulin to modifications induced by OH, causing exposure of its chromophoric aromatic amino acid residues, quenching of tyrosine fluorescence intensity, loss of α-helix and gain in β content. Modification causes re-arrangement of native interactions of the aromatic residues in insulin. It enhanced the carbonyl content in insulin, exposed its hydrophobic patches and generated non-fibrillar, amorphous type of aggregates that are cytotoxic in nature. Native insulin induced low titre antibodies in immunized rabbits, whereas OH modified insulin generated a strong immune response. Competitive ELISA studies showed high specificity of antibodies generated against OH modified insulin towards the modified protein. Cross reaction studies showed the presence of common antigenic determinants on various oxidised proteins. Since T2DM patients show increased ROS production, oxidation of insulin is expected to occur, which might amplify autoimmune reactions against insulin. True to the assumption, direct binding ELISA showed the presence of anti-OH insulin circulating antibodies in T2DM patients which are specific for the oxidized insulin. In conclusion, insulin loses structural integrity to OH, forms cytotoxic amorphous aggregates, turns highly immunogenic and elicits humoral response in T2DM patients.
Collapse
Affiliation(s)
- Mohd Talha
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Abdul Rouf Mir
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Safia Habib
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Minhal Abidi
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Mohd Sharib Warsi
- Department of Biochemistry, Jawaharlal Nehru Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Sidra Islam
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
24
|
Krzemień P, Kasperczyk S, Banach M, Kasperczyk A, Dobrakowski M, Tomasik T, Windak A, Mastej M, Catapano A, Ray KK, Mikhailidis DP, Toth PP, Howard G, Lip GY, Tomaszewski M, Charchar FJ, Sattar N, Williams B, MacDonald TM, Penson PE, Jóźwiak JJ. Serum antinuclear autoantibodies are associated with measures of oxidative stress and lifestyle factors: analysis of LIPIDOGRAM2015 and LIPIDOGEN2015 studies. Arch Med Sci 2021; 19:1214-1227. [PMID: 37732061 PMCID: PMC10507751 DOI: 10.5114/aoms/139313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 09/22/2023] Open
Abstract
Introduction Oxidative stress is one of many factors suspected to promote antinuclear autoantibody (ANA) formation. Reactive oxygen species can induce changes in the antigenic structure of macromolecules, causing the immune system to treat them as "neo-antigens" and start production of autoantibodies. This study was designed to evaluate the relationship between oxidative stress markers, lifestyle factors and the detection of ANA. Material and methods We examined measures of oxidative stress indices of free-radical damage to lipids and proteins, such as total oxidant status (TOS), concentration of protein thiol groups (PSH), and malondialdehyde (MDA), activity of superoxide dismutase (SOD) in 1731 serum samples. The parameters of the non-enzymatic antioxidant system, such as total antioxidant status (TAS) and uric acid (UA) concentration, were also measured and the oxidative stress index (OSI-index) was calculated. All samples were tested for the presence of ANA using an indirect immunofluorescence assay (IIFA). Results The presence of ANA in women was associated with lower physical activity (p = 0.036), less frequent smoking (p = 0.007) and drinking of alcohol (p = 0.024) accompanied by significant changes in SOD isoenzymes activity (p < 0.001) and a higher uric acid (UA) concentration (p < 0.001). In ANA positive males we observed lower concentrations of PSH (p = 0.046) and increased concentrations of MDA (p = 0.047). Conclusions The results indicate that local oxidative stress may be associated with increased probability of ANA formation in a sex-specific manner.
Collapse
Affiliation(s)
| | - Sławomir Kasperczyk
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice Poland
| | - Maciej Banach
- Department of Hypertension, Medical University of Lodz, Lodz, Poland
| | - Aleksandra Kasperczyk
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice Poland
| | - Michał Dobrakowski
- Department of Biochemistry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice Poland
| | - Tomasz Tomasik
- Department of Family Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Adam Windak
- Department of Family Medicine, Jagiellonian University Medical College, Krakow, Poland
| | | | - Alberico Catapano
- Department of Pharmacological Sciences, University of Milano and Multimedica IRCCS, Milano, Italy
| | - Kausik K. Ray
- Imperial Centre for Cardiovascular Disease Prevention, Department of Primary Care and Public Health, Imperial College, Kensington, London, United Kingdom
| | - Dimitri P. Mikhailidis
- Department of Clinical Biochemistry, Royal Free Hospital, University College London, London, United Kingdom
| | - Peter P. Toth
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore MD, Maryland, USA
- CGH Medical Center, Sterling, Illinois IL, USA
| | - George Howard
- Department of Biostatistics, School of Public Health of Alabama at Birmingham, Birmingham AL, USA
| | - Gregory Y.H. Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool L14 3PE, United Kingdom
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Fadi J. Charchar
- School of Health and Life Sciences, Federation University Australia, Ballarat VIC 3350, Victoria, Australia
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, United Kingdom
| | - Bryan Williams
- NIHR University College London Biomedical Research Centre, University College London and University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Thomas M. MacDonald
- MEMO Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Peter E. Penson
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
- Liverpool Centre for Cardiovascular Science, Liverpool, United Kingdom
| | - Jacek J. Jóźwiak
- Department of Family Medicine and Public Health, Faculty of Medicine, University of Opole, Opole, Poland
| |
Collapse
|
25
|
Isaguliants M, Krotova O, Petkov S, Jansons J, Bayurova E, Mezale D, Fridrihsone I, Kilpelainen A, Podschwadt P, Agapkina Y, Smirnova O, Kostic L, Saleem M, Latyshev O, Eliseeva O, Malkova A, Gorodnicheva T, Wahren B, Gordeychuk I, Starodubova E, Latanova A. Cellular Immune Response Induced by DNA Immunization of Mice with Drug Resistant Integrases of HIV-1 Clade A Offers Partial Protection against Growth and Metastatic Activity of Integrase-Expressing Adenocarcinoma Cells. Microorganisms 2021; 9:1219. [PMID: 34199989 PMCID: PMC8226624 DOI: 10.3390/microorganisms9061219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 02/07/2023] Open
Abstract
Therapeutic DNA-vaccination against drug-resistant HIV-1 may hinder emergence and spread of drug-resistant HIV-1, allowing for longer successful antiretroviral treatment (ART) up-to relief of ART. We designed DNA-vaccines against drug-resistant HIV-1 based on consensus clade A integrase (IN) resistant to raltegravir: IN_in_r1 (L74M/E92Q/V151I/N155H/G163R) or IN_in_r2 (E138K/G140S/Q148K) carrying D64V abrogating IN activity. INs, overexpressed in mammalian cells from synthetic genes, were assessed for stability, route of proteolytic degradation, and ability to induce oxidative stress. Both were found safe in immunotoxicity tests in mice, with no inherent carcinogenicity: their expression did not enhance tumorigenic or metastatic potential of adenocarcinoma 4T1 cells. DNA-immunization of mice with INs induced potent multicytokine T-cell response mainly against aa 209-239, and moderate IgG response cross-recognizing diverse IN variants. DNA-immunization with IN_in_r1 protected 60% of mice from challenge with 4Tlluc2 cells expressing non-mutated IN, while DNA-immunization with IN_in_r2 protected only 20% of mice, although tumor cells expressed IN matching the immunogen. Tumor size inversely correlated with IN-specific IFN-γ/IL-2 T-cell response. IN-expressing tumors displayed compromised metastatic activity restricted to lungs with reduced metastases size. Protective potential of IN immunogens relied on their immunogenicity for CD8+ T-cells, dependent on proteasomal processing and low level of oxidative stress.
Collapse
Affiliation(s)
- Maria Isaguliants
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden; (S.P.); (A.K.); (P.P.); (L.K.); (M.S.); (B.W.)
- Department of Research, Riga Stradins University, LV-1007 Riga, Latvia; (J.J.); (D.M.); (I.F.)
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (O.K.); (E.B.); (O.S.); (O.L.); (O.E.); (I.G.)
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia
| | - Olga Krotova
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (O.K.); (E.B.); (O.S.); (O.L.); (O.E.); (I.G.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Stefan Petkov
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden; (S.P.); (A.K.); (P.P.); (L.K.); (M.S.); (B.W.)
| | - Juris Jansons
- Department of Research, Riga Stradins University, LV-1007 Riga, Latvia; (J.J.); (D.M.); (I.F.)
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia
| | - Ekaterina Bayurova
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (O.K.); (E.B.); (O.S.); (O.L.); (O.E.); (I.G.)
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia
| | - Dzeina Mezale
- Department of Research, Riga Stradins University, LV-1007 Riga, Latvia; (J.J.); (D.M.); (I.F.)
| | - Ilze Fridrihsone
- Department of Research, Riga Stradins University, LV-1007 Riga, Latvia; (J.J.); (D.M.); (I.F.)
| | - Athina Kilpelainen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden; (S.P.); (A.K.); (P.P.); (L.K.); (M.S.); (B.W.)
| | - Philip Podschwadt
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden; (S.P.); (A.K.); (P.P.); (L.K.); (M.S.); (B.W.)
| | - Yulia Agapkina
- Department of Chemistry and Belozersky Institute of Physicochemical Biology, Moscow State University, 119991 Moscow, Russia;
| | - Olga Smirnova
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (O.K.); (E.B.); (O.S.); (O.L.); (O.E.); (I.G.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Linda Kostic
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden; (S.P.); (A.K.); (P.P.); (L.K.); (M.S.); (B.W.)
| | - Mina Saleem
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden; (S.P.); (A.K.); (P.P.); (L.K.); (M.S.); (B.W.)
| | - Oleg Latyshev
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (O.K.); (E.B.); (O.S.); (O.L.); (O.E.); (I.G.)
| | - Olesja Eliseeva
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (O.K.); (E.B.); (O.S.); (O.L.); (O.E.); (I.G.)
| | - Anastasia Malkova
- Institute of Medical Biological Research and Technologies, 143090 Krasnoznamensk, Russia;
| | | | - Britta Wahren
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden; (S.P.); (A.K.); (P.P.); (L.K.); (M.S.); (B.W.)
| | - Ilya Gordeychuk
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (O.K.); (E.B.); (O.S.); (O.L.); (O.E.); (I.G.)
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia
- Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, 127994 Moscow, Russia
| | - Elizaveta Starodubova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anastasia Latanova
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology of the Ministry of Health of the Russian Federation, 123098 Moscow, Russia; (O.K.); (E.B.); (O.S.); (O.L.); (O.E.); (I.G.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
26
|
Kehm R, Baldensperger T, Raupbach J, Höhn A. Protein oxidation - Formation mechanisms, detection and relevance as biomarkers in human diseases. Redox Biol 2021; 42:101901. [PMID: 33744200 PMCID: PMC8113053 DOI: 10.1016/j.redox.2021.101901] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/06/2021] [Accepted: 02/12/2021] [Indexed: 12/16/2022] Open
Abstract
Generation of reactive oxygen species and related oxidants is an inevitable consequence of life. Proteins are major targets for oxidation reactions, because of their rapid reaction rates with oxidants and their high abundance in cells, extracellular tissues, and body fluids. Additionally, oxidative stress is able to degrade lipids and carbohydrates to highly reactive intermediates, which eventually attack proteins at various functional sites. Consequently, a wide variety of distinct posttranslational protein modifications is formed by protein oxidation, glycoxidation, and lipoxidation. Reversible modifications are relevant in physiological processes and constitute signaling mechanisms ("redox signaling"), while non-reversible modifications may contribute to pathological situations and several diseases. A rising number of publications provide evidence for their involvement in the onset and progression of diseases as well as aging processes. Certain protein oxidation products are chemically stable and formed in large quantity, which makes them promising candidates to become biomarkers of oxidative damage. Moreover, progress in the development of detection and quantification methods facilitates analysis time and effort and contributes to their future applicability in clinical routine. The present review outlines the most important classes and selected examples of oxidative protein modifications, elucidates the chemistry beyond their formation and discusses available methods for detection and analysis. Furthermore, the relevance and potential of protein modifications as biomarkers in the context of disease and aging is summarized.
Collapse
Affiliation(s)
- Richard Kehm
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany.
| | - Tim Baldensperger
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany.
| | - Jana Raupbach
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany.
| | - Annika Höhn
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764, Muenchen-Neuherberg, Germany.
| |
Collapse
|
27
|
Lal R, Dhaliwal J, Dhaliwal N, Dharavath RN, Chopra K. Activation of the Nrf2/HO-1 signaling pathway by dimethyl fumarate ameliorates complete Freund's adjuvant-induced arthritis in rats. Eur J Pharmacol 2021; 899:174044. [PMID: 33745959 DOI: 10.1016/j.ejphar.2021.174044] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 03/08/2021] [Accepted: 03/15/2021] [Indexed: 12/13/2022]
Abstract
The nuclear factor erythroid 2-related factor (Nrf2) signaling pathway has recently emerged as a novel therapeutic target in treating various diseases. Therefore, the present study aimed to assess the protective role of the Nrf2 activator, dimethyl fumarate (DMF) in the complete Freund's adjuvant (CFA)- induced arthritis model. DMF (25, 50, and 100 mg/kg) and dexamethasone (2 mg/kg) were orally administered for 14 days. Pain-related tests, paw volume, and arthritic scores were measured weekly. Serum TNF-α, IL-1β, cyclic citrullinated peptide (CCP), C-reactive protein (CRP), and rheumatoid factor (RF) levels were estimated. Nitrite, malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase (GPx), glutathione (GSH), catalase (CAT), and myeloperoxidase (MPO) levels were also evaluated. NF-κB, Nrf2, HO-1, and COX-2 levels were estimated in the joint tissue. DMF treatment exerted anti-arthritic activity by enhancing the nociceptive threshold, improving arthritis scores, and reducing paw edema. Also, DMF suppressed changes in oxidative stress markers and inflammatory mediators and enhanced Nrf2 and HO-1 levels in CFA-injected rats. These findings indicate that the anti-arthritic activity of DMF may be mediated by the activation of the Nrf2/HO-1 pathway, which reduced oxidative damage and inflammation.
Collapse
Affiliation(s)
- Roshan Lal
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, 160014, India
| | - Jatinder Dhaliwal
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, 160014, India
| | - Navneet Dhaliwal
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, 160014, India
| | - Ravinder Naik Dharavath
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, 160014, India
| | - Kanwaljit Chopra
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
28
|
Clemen R, Freund E, Mrochen D, Miebach L, Schmidt A, Rauch BH, Lackmann J, Martens U, Wende K, Lalk M, Delcea M, Bröker BM, Bekeschus S. Gas Plasma Technology Augments Ovalbumin Immunogenicity and OT-II T Cell Activation Conferring Tumor Protection in Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003395. [PMID: 34026437 PMCID: PMC8132054 DOI: 10.1002/advs.202003395] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/22/2021] [Indexed: 05/04/2023]
Abstract
Reactive oxygen species (ROS/RNS) are produced during inflammation and elicit protein modifications, but the immunological consequences are largely unknown. Gas plasma technology capable of generating an unmatched variety of ROS/RNS is deployed to mimic inflammation and study the significance of ROS/RNS modifications using the model protein chicken ovalbumin (Ova vs oxOva). Dynamic light scattering and circular dichroism spectroscopy reveal structural modifications in oxOva compared to Ova. T cells from Ova-specific OT-II but not from C57BL/6 or SKH-1 wild type mice presents enhanced activation after Ova addition. OxOva exacerbates this activation when administered ex vivo or in vivo, along with an increased interferon-gamma production, a known anti-melanoma agent. OxOva vaccination of wild type mice followed by inoculation of syngeneic B16F10 Ova-expressing melanoma cells shows enhanced T cell number and activation, decreased tumor burden, and elevated numbers of antigen-presenting cells when compared to their Ova-vaccinated counterparts. Analysis of oxOva using mass spectrometry identifies three hot spots regions rich in oxidative modifications that are associated with the increased T cell activation. Using Ova as a model protein, the findings suggest an immunomodulating role of multi-ROS/RNS modifications that may spur novel research lines in inflammation research and for vaccination strategies in oncology.
Collapse
Affiliation(s)
- Ramona Clemen
- ZIK plasmatisLeibniz Institute for Plasma Science and Technology (INP)Felix‐Hausdorff‐Str. 2Greifswald17489Germany
| | - Eric Freund
- ZIK plasmatisLeibniz Institute for Plasma Science and Technology (INP)Felix‐Hausdorff‐Str. 2Greifswald17489Germany
- Department of GeneralVisceralThoracicand Vascular SurgeryUniversity Medicine GreifswaldSauerbruchstr. DZ7Greifswald17475Germany
| | - Daniel Mrochen
- ZIK plasmatisLeibniz Institute for Plasma Science and Technology (INP)Felix‐Hausdorff‐Str. 2Greifswald17489Germany
- Department of ImmunologyUniversity Medicine GreifswaldSauerbruchstr. DZ7Greifswald17475Germany
| | - Lea Miebach
- ZIK plasmatisLeibniz Institute for Plasma Science and Technology (INP)Felix‐Hausdorff‐Str. 2Greifswald17489Germany
- Department of GeneralVisceralThoracicand Vascular SurgeryUniversity Medicine GreifswaldSauerbruchstr. DZ7Greifswald17475Germany
| | - Anke Schmidt
- ZIK plasmatisLeibniz Institute for Plasma Science and Technology (INP)Felix‐Hausdorff‐Str. 2Greifswald17489Germany
| | - Bernhard H. Rauch
- Institute of Pharmacology (C_Dat)University Medicine GreifswaldFelix‐Hausdorff‐Str. 1Greifswald17489Germany
| | - Jan‐Wilm Lackmann
- CECAD proteomics facilityUniversity of CologneJoseph‐Stelzmann‐Str. 26Cologne50931Germany
| | - Ulrike Martens
- ZIK HIKEUniversity of GreifswaldFleischmannstr. 42–44Greifswald17489Germany
- Institute of BiochemistryUniversity of GreifswaldFelix‐Hausdorff‐Str. 4Greifswald17489Germany
| | - Kristian Wende
- ZIK plasmatisLeibniz Institute for Plasma Science and Technology (INP)Felix‐Hausdorff‐Str. 2Greifswald17489Germany
| | - Michael Lalk
- Institute of BiochemistryUniversity of GreifswaldFelix‐Hausdorff‐Str. 4Greifswald17489Germany
| | - Mihaela Delcea
- ZIK HIKEUniversity of GreifswaldFleischmannstr. 42–44Greifswald17489Germany
- Institute of BiochemistryUniversity of GreifswaldFelix‐Hausdorff‐Str. 4Greifswald17489Germany
| | - Barbara M. Bröker
- Department of ImmunologyUniversity Medicine GreifswaldSauerbruchstr. DZ7Greifswald17475Germany
| | - Sander Bekeschus
- ZIK plasmatisLeibniz Institute for Plasma Science and Technology (INP)Felix‐Hausdorff‐Str. 2Greifswald17489Germany
| |
Collapse
|
29
|
Lou A, Wang L, Lai W, Zhu D, Wu W, Wang Z, Cai Z, Yang M. Advanced oxidation protein products induce inflammatory responses and invasive behaviour in fibroblast-like synoviocytes via the RAGE-NF-κB pathway. Bone Joint Res 2021; 10:259-268. [PMID: 33827262 PMCID: PMC8077182 DOI: 10.1302/2046-3758.104.bjr-2020-0085.r2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aims Rheumatoid arthritis (RA), which mainly results from fibroblast-like synoviocyte (FLS) dysfunction, is related to oxidative stress. Advanced oxidation protein products (AOPPs), which are proinflammatory mediators and a novel biomarker of oxidative stress, have been observed to accumulate significantly in the serum of RA patients. Here, we present the first investigation of the effects of AOPPs on RA-FLSs and the signalling pathway involved in AOPP-induced inflammatory responses and invasive behaviour. Methods We used different concentrations of AOPPs (50 to 200 µg/ml) to treat RA-FLSs. Cell migration and invasion and the expression levels of tumour necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), matrix metalloproteinase-3 (MMP-3), and MMP-13 were investigated. Western blot and immunofluorescence were used to analyze nuclear factor-κB (NF-κB) activation. Results AOPPs promoted RA-FLS migration and invasion in vitro and significantly induced the messenger RNA (mRNA) and protein expression of TNF-α, IL-6, MMP-3, and MMP-13 in dose- and time-dependent manners. Moreover, AOPPs markedly activated the phosphorylation of nuclear factor-κB (NF-κB) p65 protein, which triggered inhibitory kappa B-alpha (IκBα) degradation, NF-κB p65 protein phosphorylation, and NF-κB p65 translocation into the nucleus. Furthermore, treatment with a neutralizing antibody specific to receptor for advanced glycation end products (RAGE) significantly suppressed aggressive behaviour and inflammation, decreased TNF-α, IL-6, MMP-3, and MMP-13 expression, and blocked AOPP-induced NF-κB pathway activation. Conclusion The results indicate that AOPPs can enhance aggressive behaviour and the inflammatory response in RA-FLSs via the RAGE–NF-κB pathway. These results present AOPPs as a new class of potentially important mediators of progressive disease in RA patients. Cite this article: Bone Joint Res 2021;10(4):259–268.
Collapse
Affiliation(s)
- Aiju Lou
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangdong, China.,Department of Rheumatology and Immunology, Liwan Central Hospital of Guangzhou, Guangzhou, Guangdong, China
| | - Le Wang
- Department of Orthopaedic Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,Medical Technology and Related Equipment Research for Spinal Injury Treatment, City Key Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Weinan Lai
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangdong, China
| | - DingJi Zhu
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangdong, China
| | - Weirong Wu
- Department of Rheumatology and Immunology, Liwan Central Hospital of Guangzhou, Guangzhou, Guangdong, China
| | - Zhao Wang
- Department of Orthopaedic Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,Medical Technology and Related Equipment Research for Spinal Injury Treatment, City Key Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zihong Cai
- Department of Orthopaedic Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,Medical Technology and Related Equipment Research for Spinal Injury Treatment, City Key Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Min Yang
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangdong, China
| |
Collapse
|
30
|
Dyakin VV, Wisniewski TM, Lajtha A. Racemization in Post-Translational Modifications Relevance to Protein Aging, Aggregation and Neurodegeneration: Tip of the Iceberg. Symmetry (Basel) 2021; 13:455. [PMID: 34350031 PMCID: PMC8330555 DOI: 10.3390/sym13030455] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Homochirality of DNA and prevalent chirality of free and protein-bound amino acids in a living organism represents the challenge for modern biochemistry and neuroscience. The idea of an association between age-related disease, neurodegeneration, and racemization originated from the studies of fossils and cataract disease. Under the pressure of new results, this concept has a broader significance linking protein folding, aggregation, and disfunction to an organism's cognitive and behavioral functions. The integrity of cognitive function is provided by a delicate balance between the evolutionarily imposed molecular homo-chirality and the epigenetic/developmental impact of spontaneous and enzymatic racemization. The chirality of amino acids is the crucial player in the modulation the structure and function of proteins, lipids, and DNA. The collapse of homochirality by racemization is the result of the conformational phase transition. The racemization of protein-bound amino acids (spontaneous and enzymatic) occurs through thermal activation over the energy barrier or by the tunnel transfer effect under the energy barrier. The phase transition is achieved through the intermediate state, where the chirality of alpha carbon vanished. From a thermodynamic consideration, the system in the homo-chiral (single enantiomeric) state is characterized by a decreased level of entropy. The oscillating protein chirality is suggesting its distinct significance in the neurotransmission and flow of perceptual information, adaptive associative learning, and cognitive laterality. The common pathological hallmarks of neurodegenerative disorders include protein misfolding, aging, and the deposition of protease-resistant protein aggregates. Each of the landmarks is influenced by racemization. The brain region, cell type, and age-dependent racemization critically influence the functions of many intracellular, membrane-bound, and extracellular proteins including amyloid precursor protein (APP), TAU, PrP, Huntingtin, α-synuclein, myelin basic protein (MBP), and collagen. The amyloid cascade hypothesis in Alzheimer's disease (AD) coexists with the failure of amyloid beta (Aβ) targeting drug therapy. According to our view, racemization should be considered as a critical factor of protein conformation with the potential for inducing order, disorder, misfolding, aggregation, toxicity, and malfunctions.
Collapse
Affiliation(s)
- Victor V. Dyakin
- Virtual Reality Perception Lab (VRPL), The Nathan S. Kline Institute for Psychiatric Research (NKI), Orangeburg, NY 10962, USA
| | - Thomas M. Wisniewski
- Departments of Neurology, Pathology and Psychiatry, Center for Cognitive Neurology, New York University School of Medicine, New York, NY 10016, USA
| | - Abel Lajtha
- Center for Neurochemistry, The Nathan S. Kline Institute for Psychiatric Research (NKI), Orangeburg, NY 10962, USA
| |
Collapse
|
31
|
Zia A, Farkhondeh T, Pourbagher-Shahri AM, Samarghandian S. The Roles of mitochondrial dysfunction and Reactive Oxygen Species in Aging and Senescence. Curr Mol Med 2021; 22:37-49. [PMID: 33602082 DOI: 10.2174/1566524021666210218112616] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/16/2020] [Accepted: 12/23/2020] [Indexed: 11/22/2022]
Abstract
The aging process deteriorates organs' function at different levels, causing its progressive decline to resist stress, damage, and disease. In addition to alterations in metabolic control and gene expression, the rate of aging has been connected with the generation of high amounts of Reactive Oxygen Species (ROS). The essential perspective in free radical biology is that reactive oxygen species (ROS) and free radicals are toxic, mostly cause direct biological damage to targets, and are thus a major cause of oxidative stress. Different enzymatic and non-enzymatic compounds in the cells have roles in neutralizing this toxicity. Oxidative damage in aging is mostly high in particular molecular targets, such as mitochondrial DNA and aconitase, and oxidative stress in mitochondria can cause tissue aging across intrinsic apoptosis. Mitochondria's function and morphology are impaired through aging, following a decrease in the membrane potential by an increase in peroxide generation and size of the organelles. Telomeres may be the significant trigger of replicative senescence. Oxidative stress accelerates telomere loss, whereas antioxidants slow it down. Oxidative stress is a crucial modulator of telomere shortening, and that telomere-driven replicative senescence is mainly a stress response. The age-linked mitochondrial DNA mutation and protein dysfunction aggregate in some organs like the brain and skeletal muscle, thus contributing considerably to these post-mitotic tissues' aging. The aging process is mostly due to accumulated damage done by harmful species in some macromolecules such proteins, DNA, and lipids. The degradation of non-functional, oxidized proteins is a crucial part of the antioxidant defenses of cells, in which the clearance of these proteins occurs through autophagy in the cells, which is known as mitophagy for mitochondria.
Collapse
Affiliation(s)
- Aliabbas Zia
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran. Iran
| | - Tahereh Farkhondeh
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand. Iran
| | | | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur. Iran
| |
Collapse
|
32
|
Sakata K, Yasuoka H, Yoshimoto K, Takeuchi T. Decreased activation of ataxia telangiectasia mutated (ATM) in monocytes from patients with systemic sclerosis. Rheumatology (Oxford) 2021; 59:3961-3970. [PMID: 32743653 DOI: 10.1093/rheumatology/keaa312] [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: 12/24/2019] [Revised: 03/06/2020] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES The regulation system for oxidative stress in systemic sclerosis (SSc) remains unclear. This study aimed to clarify the possible involvement of ataxia telangiectasia mutated (ATM), which plays a key role in DNA repair and redox balance, in the pathogenesis of SSc. METHODS Thirty patients with SSc and 15 healthy controls were enrolled. Expression of ATM and phosphorylated ATM (pATM), an activated form of ATM, in phagocytes in whole blood samples was analysed by FACS. Correlations between expression levels of ATM/pATM and clinical parameters of SSc patients were statistically analysed. Peripheral monocytes were cultured with an ATM-specific inhibitor (KU55933), and reactive oxygen species production in the cells was measured. RESULTS Expression level of pATM in peripheral monocytes and neutrophils from SSc patients was significantly lower than those in healthy controls (P = 0.04 and P < 0.001, respectively), while no significant difference in total ATM expression was observed between SSc and healthy controls. In addition, pATM expression in monocytes of SSc patients with interstitial lung disease or digital pitting scar was remarkably lower than in the patients without these clinical features (P = 0.02 and P = 0.03), respectively. Moreover, pATM expression in monocytes positively correlated with forced vital capacity and negatively correlated with the serum Krebs von den Lungen-6 level. Notably, KU55933, an ATM-specific inhibitor, enhanced reactive oxygen species production by monocytes under oxidative stress. CONCLUSION Our data revealed that decreased ATM activation in monocytes was associated with SSc-interstitial lung disease and that impaired ATM activation in monocytes may contribute to the disease process of SSc via uncontrolled reactive oxygen species production.
Collapse
Affiliation(s)
- Komei Sakata
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo
| | - Hidekata Yasuoka
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo.,Division of Infectious Diseases and Rheumatology, Department of Internal Medicine, Fujita Health University School of Medicine, Aichi
| | - Keiko Yoshimoto
- Department of Biotechnology, Tokyo Technical College, Tokyo, Japan
| | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo
| |
Collapse
|
33
|
Ramani S, Pathak A, Dalal V, Paul A, Biswas S. Oxidative Stress in Autoimmune Diseases: An Under Dealt Malice. Curr Protein Pept Sci 2021; 21:611-621. [PMID: 32056521 DOI: 10.2174/1389203721666200214111816] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 12/27/2022]
Abstract
Oxidative stress is the off-balance of antioxidants and free radicals. All kinds of diseases and disorders give rise to oxidative damage including autoimmune diseases. An autoimmune disorder is a pathological condition characterized by the breakdown of self-tolerance of the immune system in the body. Immunological processes against tissues and organs lead to enhanced oxidative stress and, in turn, misbalance of oxidative stress aggravates the pathobiology of the disease. Highly reactive nature of free radicals, for example hydroxyl and superoxide ions, alters DNA, protein, and lipids in the body which augment the pathologic processes of diseases. The damaged biomolecules are responsible for systemic complications and secondary disease co-morbidities. In this review, we discuss the role of oxidative stress in some incapacitating autoimmune diseases like Rheumatoid arthritis, Systemic Lupus Erythematosus, Type 1 Diabetes, and Multiple Sclerosis. Oxidative stress plays a central and course defining role in these diseases and it has become a necessity to study the pathological mechanism involved in oxidative stress to better understand and offer treatment holistically. Presently there are no clinically available parameters for measurement and treatment of pathological oxidative stress, therefore it requires intensive research. Probably, in the future, the discovery of easily detectable markers of oxidative stress can aid in the diagnosis, prognosis, and treatment of progressively destructive autoimmune diseases.
Collapse
Affiliation(s)
- Sheetal Ramani
- Department of Integrative and Functional Genomics, CSIR- Institute of Genomics and Integrative Biology, New Delhi, 110007, India
| | - Ayush Pathak
- Department of Integrative and Functional Genomics, CSIR- Institute of Genomics and Integrative Biology, New Delhi, 110007, India
| | - Vikram Dalal
- Department of Biotechnology, Indian Institute of Technology, Roorkee, 247667, India
| | - Anamika Paul
- School of Engineering and Technology, Ansal University, Gurugram, Haryana, 122003, India
| | - Sagarika Biswas
- Department of Integrative and Functional Genomics, CSIR- Institute of Genomics and Integrative Biology, New Delhi, 110007, India
| |
Collapse
|
34
|
Atukorala I, Mathivanan S. The Role of Post-Translational Modifications in Targeting Protein Cargo to Extracellular Vesicles. Subcell Biochem 2021; 97:45-60. [PMID: 33779913 DOI: 10.1007/978-3-030-67171-6_3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Extracellular vesicles (EVs) are naturally occurring nanoparticles that contain proteins and nucleic acids. It is speculated that cells release EVs loaded with a selective cargo of proteins through highly regulated processes. Several proteomic and biochemical studies have highlighted phosphorylated, glycosylated, ubiquitinated, SUMOylated, oxidated and palmitoylated proteins within the EVs. Emerging evidences suggest that post-translational modifications (PTMs) can regulate the sorting of specific proteins into EVs and such proteins with specific PTMs have also been identified in clinical samples. Hence, it has been proposed that EV proteins with PTMs could be used as potential biomarkers of disease conditions. Among the other cellular mechanisms, the endosomal sorting complex required for transport (ESCRT) is also implicated in cargo sorting into EVs. In this chapter, various PTMs that are shown to regulate protein cargo sorting into EVs will be discussed.
Collapse
Affiliation(s)
- Ishara Atukorala
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia.
| | - Suresh Mathivanan
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
35
|
Fok JA, Mayer C. Genetic-Code-Expansion Strategies for Vaccine Development. Chembiochem 2020; 21:3291-3300. [PMID: 32608153 PMCID: PMC7361271 DOI: 10.1002/cbic.202000343] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/29/2020] [Indexed: 12/16/2022]
Abstract
By providing long-term protection against infectious diseases, vaccinations have significantly reduced death and morbidity worldwide. In the 21st century, (bio)technological advances have paved the way for developing prophylactic vaccines that are safer and more effective as well as enabling the use of vaccines as therapeutics to treat human diseases. Here, we provide a focused review of the utility of genetic code expansion as an emerging tool for the development of vaccines. Specifically, we discuss how the incorporation of immunogenic noncanonical amino acids can aid in eliciting immune responses against adverse self-proteins and highlight the potential of an expanded genetic code for the construction of replication-incompetent viruses. We close the review by discussing the future prospects and remaining challenges for the application of these approaches in the development of both prophylactic and therapeutic vaccines in the near future.
Collapse
Affiliation(s)
- Jelle A. Fok
- Stratingh Institute for ChemistryUniversity of GroningenNijenborgh 49474 AGGroningen (TheNetherlands
| | - Clemens Mayer
- Stratingh Institute for ChemistryUniversity of GroningenNijenborgh 49474 AGGroningen (TheNetherlands
| |
Collapse
|
36
|
Abate G, Vezzoli M, Sandri M, Rungratanawanich W, Memo M, Uberti D. Mitochondria and cellular redox state on the route from ageing to Alzheimer's disease. Mech Ageing Dev 2020; 192:111385. [PMID: 33129798 DOI: 10.1016/j.mad.2020.111385] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/05/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023]
Abstract
Several theories have been postulated, trying to explain why and how living organisms age. Despite some controversies and still huge open questions, a growing body of evidence suggest alterations of mitochondrial functionality and redox-homeostasis occur during the ageing process. Oxidative damage and mitochondrial dysfunction do not represent the cause of ageing per se but they have to be analyzed within the complexity of those series of processes occurring during lifespan. The establishment of a crosstalk among them is a shared common feature of many chronic age-related diseases, including neurodegenerative disorders, for which ageing is a major risk factor. The challenge is to understand when and how the interplay between these two systems move towards from normal ageing process to a pathological phenotype. Here in this review, we discuss the crosstalk between mitochondria and cytosolic-ROS. Furthermore, through a visual data mining approach, we attempt to describe the dynamic interplay between mitochondria and cellular redox state on the route from ageing to an AD phenotype.
Collapse
Affiliation(s)
- G Abate
- Department of Molecular and Translational Medicine, University of Brescia, Italy.
| | - M Vezzoli
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - M Sandri
- Big & Open Data Innovation Laboratory (BODaI-Lab), Department of Economics and Management, University of Brescia, Italy
| | - W Rungratanawanich
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - M Memo
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - D Uberti
- Department of Molecular and Translational Medicine, University of Brescia, Italy; Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| |
Collapse
|
37
|
The Immunopathology of Complement Proteins and Innate Immunity in Autoimmune Disease. Clin Rev Allergy Immunol 2020; 58:229-251. [PMID: 31834594 DOI: 10.1007/s12016-019-08774-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The complement is a powerful cascade of the innate immunity and also acts as a bridge between innate and acquired immune defence. Complement activation can occur via three distinct pathways, the classical, alternative and lectin pathways, each resulting in the common terminal pathway. Complement activation results in the release of a range of biologically active molecules that significantly contribute to immune surveillance and tissue homeostasis. Several soluble and membrane-bound regulatory proteins restrict complement activation in order to prevent complement-mediated autologous damage, consumption and exacerbated inflammation. The crucial role of complement in the host homeostasis is illustrated by association of both complement deficiency and overactivation with severe and life-threatening diseases. Autoantibodies targeting complement components have been described to alter expression and/or function of target protein resulting in a dysregulation of the delicate equilibrium between activation and inhibition of complement. The spectrum of diseases associated with complement autoantibodies depends on which complement protein and activation pathway are targeted, ranging from autoimmune disorders to kidney and vascular diseases. Nevertheless, these autoantibodies have been identified as differential biomarkers for diagnosis or follow-up of disease only in a small number of clinical conditions. For some autoantibodies, a clear relationship with clinical manifestations has been identified, such as anti-C1q, anti-Factor H, anti-C1 Inhibitor antibodies and C3 nephritic factor. For other autoantibodies, the origin and the functional consequences still remain to be elucidated, questioning about the pathophysiological significance of these autoantibodies, such as anti-mannose binding lectin, anti-Factor I, anti-Factor B and anti-C3b antibodies. The detection of autoantibodies targeting complement components is performed in specialized laboratories; however, there is no consensus on detection methods and standardization of the assays is a real challenge. This review summarizes the current panorama of autoantibodies targeting complement recognition proteins of the classical and lectin pathways, associated proteases, convertases, regulators and terminal components, with an emphasis on autoantibodies clearly involved in clinical conditions.
Collapse
|
38
|
Finelli MJ. Redox Post-translational Modifications of Protein Thiols in Brain Aging and Neurodegenerative Conditions-Focus on S-Nitrosation. Front Aging Neurosci 2020; 12:254. [PMID: 33088270 PMCID: PMC7497228 DOI: 10.3389/fnagi.2020.00254] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/24/2020] [Indexed: 12/14/2022] Open
Abstract
Reactive oxygen species and reactive nitrogen species (RONS) are by-products of aerobic metabolism. RONS trigger a signaling cascade that can be transduced through oxidation-reduction (redox)-based post-translational modifications (redox PTMs) of protein thiols. This redox signaling is essential for normal cellular physiology and coordinately regulates the function of redox-sensitive proteins. It plays a particularly important role in the brain, which is a major producer of RONS. Aberrant redox PTMs of protein thiols can impair protein function and are associated with several diseases. This mini review article aims to evaluate the role of redox PTMs of protein thiols, in particular S-nitrosation, in brain aging, and in neurodegenerative diseases. It also discusses the potential of using redox-based therapeutic approaches for neurodegenerative conditions.
Collapse
Affiliation(s)
- Mattéa J Finelli
- School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
39
|
Redox Regulation of PPAR γ in Polarized Macrophages. PPAR Res 2020; 2020:8253831. [PMID: 32695149 PMCID: PMC7350077 DOI: 10.1155/2020/8253831] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 04/28/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022] Open
Abstract
The peroxisome proliferator-activated receptor (PPARγ) is a central mediator of cellular lipid metabolism and immune cell responses during inflammation. This is facilitated by its role as a transcription factor as well as a DNA-independent protein interaction partner. We addressed how the cellular redox milieu in the cytosol and the nucleus of lipopolysaccharide (LPS)/interferon-γ- (IFNγ-) and interleukin-4- (IL4-) polarized macrophages (MΦ) initiates posttranslational modifications of PPARγ, that in turn alter its protein function. Using the redox-sensitive GFP2 (roGFP2), we validated oxidizing and reducing conditions following classical and alternative activation of MΦ, while the redox status of PPARγ was determined via mass spectrometry. Cysteine residues located in the zinc finger regions (amino acid fragments AA 90-115, AA 116-130, and AA 160-167) of PPARγ were highly oxidized, accompanied by phosphorylation of serine 82 in response to LPS/IFNγ, whereas IL4-stimulation provoked minor serine 82 phosphorylation and less cysteine oxidation, favoring a reductive milieu. Mutating these cysteines to alanine to mimic a redox modification decreased PPARγ-dependent reporter gene transactivation supporting a functional shift of PPARγ associated with the MΦ phenotype. These data suggest distinct mechanisms for regulating PPARγ function based on the redox state of MΦ.
Collapse
|
40
|
Glencross DA, Ho TR, Camiña N, Hawrylowicz CM, Pfeffer PE. Air pollution and its effects on the immune system. Free Radic Biol Med 2020; 151:56-68. [PMID: 32007522 DOI: 10.1016/j.freeradbiomed.2020.01.179] [Citation(s) in RCA: 290] [Impact Index Per Article: 72.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 12/22/2022]
Abstract
A well-functioning immune system is vital for a healthy body. Inadequate and excessive immune responses underlie diverse pathologies such as serious infections, metastatic malignancies and auto-immune conditions. Therefore, understanding the effects of ambient pollutants on the immune system is vital to understanding how pollution causes disease, and how that pathology could be abrogated. The immune system itself consists of multiple types of immune cell that act together to generate (or fail to generate) immune responses and in this article we review evidence of how air pollutants can affect different immune cell types such as particle-clearing macrophages, inflammatory neutrophils, dendritic cells that orchestrate adaptive immune responses and lymphocytes that enact those responses. Common themes that emerge are of the capacity of air pollutants to stimulate pro-inflammatory immune responses across multiple classes of immune cell. Air pollution can enhance T helper lymphocyte type 2 (Th2) and T helper lymphocyte type 17 (Th17) adaptive immune responses, as seen in allergy and asthma, and dysregulate anti-viral immune responses. The clinical effects of air pollution, in particular the known association between elevated ambient pollution and exacerbations of asthma and chronic obstructive pulmonary disease (COPD), are consistent with these identified immunological mechanisms. Further to this, as inhaled air pollution deposits primarily on the respiratory mucosa this review focuses on mechanisms of respiratory disease. However, as discussed in the article, air pollution also affects the wider immune system for example in the neonate and gastrointestinal tract. Whilst the many identified actions of air pollution on the immune system are notably diverse, immunological research does suggest potential strategies to ameliorate such effects, for example with vitamin D supplementation. An in-depth understanding of the immunological effects of ambient pollutants should hopefully yield new ideas on how to reduce the adverse health effects of air pollution.
Collapse
Affiliation(s)
- Drew A Glencross
- Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, Guy's Hospital, London, SE1 9RT, UK; MRC Centre for Environment and Health, King's College London, Franklin Wilkins Building, London, SE1 9NH, UK
| | - Tzer-Ren Ho
- Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, Guy's Hospital, London, SE1 9RT, UK; MRC Centre for Environment and Health, King's College London, Franklin Wilkins Building, London, SE1 9NH, UK
| | - Nuria Camiña
- MRC Centre for Environment and Health, King's College London, Franklin Wilkins Building, London, SE1 9NH, UK
| | - Catherine M Hawrylowicz
- Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, Guy's Hospital, London, SE1 9RT, UK.
| | - Paul E Pfeffer
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| |
Collapse
|
41
|
Vinci C, Infantino M, Raturi S, Tindell A, Topping LM, Strollo R, Amital H, Shoenfeld Y, Gertel S, Grossi V, Manfredi M, Rutigliano IM, Bandinelli F, Li Gobbi F, Damiani A, Pozzilli P, Mcinnes IB, Goodyear CS, Benucci M, Nissim A. Immunoglobulin A antibodies to oxidized collagen type II as a potential biomarker for the stratification of spondyloarthritis from rheumatoid arthritis. Scand J Rheumatol 2020; 49:281-291. [PMID: 32314641 DOI: 10.1080/03009742.2020.1713395] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVES The discovery of diseased tissue-specific neoantigens offers the opportunity to develop important disease tissue-specific biomarkers that can help in the prediction, diagnosis, and stratification of diseases. This opportunity is specifically significant for autoimmune diseases where diagnostic biomarkers are not available. Inflammatory autoimmune diseases are commonly associated with local generation of large amounts of reactive oxidants. We have previously identified oxidative post-translationally modified (oxPTM) tissue-specific neoantigens in rheumatoid arthritis (RA) and type 1 diabetes that elicit an immune response. In the current study, we studied the presence and clinical significance of antibodies to oxPTM collagen type II (CII) in patients with spondyloarthritis (SpA). METHOD Levels of antibodies specific to native CII and oxPTM-CII were assessed by enzyme-linked immunosorbent assay. RESULTS Immunoglobulin G (IgG) binding to oxPTM-CII was observed in 52%, 83%, and 28% of serum samples from patients with axial spondyloarthritis (axSpA), RA, and psoriatic arthritis (PsA), respectively. Importantly, while strong IgA anti-oxPTM-CII responses were detected in axSpA and PsA patients, with 47% and 84% respective binders, no IgA anti-oxPTM-CII was detected in RA patients. IgA anti-oxPTM-CII reactivity in axSpA patients treated with biologics was higher and more frequent, with 85% binders compared to 9% binders in patients treated with synthetic disease-modifying anti-rheumatic drugs. CONCLUSION Our data imply that SpA and PsA are associated with the presence of antibodies to oxPTM-CII, suggesting that there may be a humoral component that may distinguish patients with SpA from RA. Our approach could be adapted to other diseases, particularly to inflammatory autoimmune diseases.
Collapse
Affiliation(s)
- C Vinci
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London , London, UK.,Department of Endocrinology and Diabetes, Campus Biomedico , Rome, Italy
| | - M Infantino
- Immunology and Allergology Laboratory Unit, S.Giovanni di Dio Hospital , Florence, Italy
| | - S Raturi
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London , London, UK
| | - A Tindell
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow , Glasgow, UK
| | - L M Topping
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London , London, UK
| | - R Strollo
- Department of Endocrinology and Diabetes, Campus Biomedico , Rome, Italy
| | - H Amital
- Department of Internal Medicine B and Zabludowicz Center for Autoimmune Diseases, Sheba Medical Centre , Ramat Gan, Israel
| | - Y Shoenfeld
- Department of Internal Medicine B and Zabludowicz Center for Autoimmune Diseases, Sheba Medical Centre , Ramat Gan, Israel
| | - S Gertel
- Department of Internal Medicine B and Zabludowicz Center for Autoimmune Diseases, Sheba Medical Centre , Ramat Gan, Israel
| | - V Grossi
- Immunology and Allergology Laboratory Unit, S.Giovanni di Dio Hospital , Florence, Italy
| | - M Manfredi
- Immunology and Allergology Laboratory Unit, S.Giovanni di Dio Hospital , Florence, Italy
| | - I M Rutigliano
- Rheumatology Unit, Sab.Giovanni di Dio Hospital , Florence, Italy
| | - F Bandinelli
- Rheumatology Unit, Sab.Giovanni di Dio Hospital , Florence, Italy
| | - F Li Gobbi
- Rheumatology Unit, Sab.Giovanni di Dio Hospital , Florence, Italy
| | - A Damiani
- Rheumatology Unit, Sab.Giovanni di Dio Hospital , Florence, Italy
| | - P Pozzilli
- Department of Endocrinology and Diabetes, Campus Biomedico , Rome, Italy
| | - I B Mcinnes
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow , Glasgow, UK
| | - C S Goodyear
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow , Glasgow, UK
| | - M Benucci
- Rheumatology Unit, Sab.Giovanni di Dio Hospital , Florence, Italy
| | - A Nissim
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London , London, UK
| |
Collapse
|
42
|
Ji H, Wu G, Li Y, Wang K, Xue X, You S, Wu S, Ren T, He B, Shi X. Self-Albumin Camouflage of Carrier Protein Prevents Nontarget Antibody Production for Enhanced LDL-C Immunotherapy. Adv Healthc Mater 2020; 9:e1901203. [PMID: 31814301 DOI: 10.1002/adhm.201901203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/07/2019] [Indexed: 11/05/2022]
Abstract
Elevated low-density lipoprotein cholesterol (LDL-C) increases the risk of atherosclerotic cardiovascular disease. Peptide-based PCSK9 vaccines have shown a promising prospect of reducing LDL-C. In peptide vaccine (pVax) design, the peptide antigens need to conjugate with carrier protein (CP). However, CP incorporation can induce undesirable anti-CP antibodies, which sterically mask peptide epitopes from being recognized by specific B cells and impair subsequent therapeutically antibody production. This epitopic suppression has posed a barrier in clinical translation of conjugate vaccines all along. A model CP (keyhole limpet hemocyanin, KLH) is herein camouflaged with serum albumin (SA) into hybrid nanocarriers (SA@N), with PCSK9 peptide being anchored onto the surface to form nanovaccine (SA@NVax). Such camouflage of KLH via high "self" SA coverage is able to inhibit KLH from extracellular immune recognition and prevent detectable anti-KLH antibody production. Furthermore, the nanovaccine around 70 nm stabilized by intermolecular disulfide network is ideal for internalization and biodegradation by antigen presenting cells as well as better retention in draining lymph nodes and spleen. As expected, the SA@NVax efficiently primes higher anti-PCSK9 IgG antibody titer than PCSK9 pVax.
Collapse
Affiliation(s)
- Haiying Ji
- Department of Anesthesiology and SICUXinhua HospitalSchool of MedicineShanghai Jiao Tong University Shanghai 200092 China
| | - Guangxi Wu
- Department of Anesthesiology and SICUXinhua HospitalSchool of MedicineShanghai Jiao Tong University Shanghai 200092 China
| | - Yongyong Li
- Institute for Biomedical Engineering and Nano ScienceShanghai East HospitalTongji University School of Medicine Shanghai 200092 China
| | - Kun Wang
- Institute for Biomedical Engineering and Nano ScienceShanghai East HospitalTongji University School of Medicine Shanghai 200092 China
| | - Xiaomei Xue
- Department of Anesthesiology and SICUXinhua HospitalSchool of MedicineShanghai Jiao Tong University Shanghai 200092 China
| | - Shasha You
- Department of Anesthesiology and SICUXinhua HospitalSchool of MedicineShanghai Jiao Tong University Shanghai 200092 China
| | - Shengming Wu
- Institute for Biomedical Engineering and Nano ScienceShanghai East HospitalTongji University School of Medicine Shanghai 200092 China
| | - Tianbin Ren
- School of Materials Science and EngineeringTongji University Shanghai 200092 China
| | - Bin He
- Department of Anesthesiology and SICUXinhua HospitalSchool of MedicineShanghai Jiao Tong University Shanghai 200092 China
| | - Xueyin Shi
- Department of Anesthesiology and SICUXinhua HospitalSchool of MedicineShanghai Jiao Tong University Shanghai 200092 China
| |
Collapse
|
43
|
Ren J, Catalina MD, Eden K, Liao X, Read KA, Luo X, McMillan RP, Hulver MW, Jarpe M, Bachali P, Grammer AC, Lipsky PE, Reilly CM. Selective Histone Deacetylase 6 Inhibition Normalizes B Cell Activation and Germinal Center Formation in a Model of Systemic Lupus Erythematosus. Front Immunol 2019; 10:2512. [PMID: 31708928 PMCID: PMC6823248 DOI: 10.3389/fimmu.2019.02512] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 10/08/2019] [Indexed: 01/25/2023] Open
Abstract
Autoantibody production by plasma cells (PCs) plays a pivotal role in the pathogenesis of systemic lupus erythematosus (SLE). The molecular pathways by which B cells become pathogenic PC secreting autoantibodies in SLE are incompletely characterized. Histone deactylase 6 (HDAC6) is a unique cytoplasmic HDAC that modifies the interaction of a number of tubulin- associated proteins; inhibition of HDAC6 has been shown to be beneficial in murine models of SLE, but the downstream pathways accounting for the therapeutic benefit have not been clearly delineated. In the current study, we sought to determine whether selective HDAC6 inhibition would abrogate abnormal B cell activation in SLE. We treated NZB/W lupus mice with the selective HDAC6 inhibitor, ACY-738, for 4 weeks beginning at 20 weeks-of age. After only 4 weeks of treatment, manifestation of lupus nephritis (LN) were greatly reduced in these animals. We then used RNAseq to determine the genomic signatures of splenocytes from treated and untreated mice and applied computational cellular and pathway analysis to reveal multiple signaling events associated with B cell activation and differentiation in SLE that were modulated by HDAC6 inhibition. PC development was abrogated and germinal center (GC) formation was greatly reduced. When the HDAC6 inhibitor-treated lupus mouse gene signatures were compared to human lupus patient gene signatures, the results showed numerous immune, and inflammatory pathways increased in active human lupus were significantly decreased in the HDAC6 inhibitor treated animals. Pathway analysis suggested alterations in cellular metabolism might contribute to the normalization of lupus mouse spleen genomic signatures, and this was confirmed by direct measurement of the impact of the HDAC6 inhibitor on metabolic activities of murine spleen cells. Taken together, these studies show HDAC6 inhibition decreases B cell activation signaling pathways and reduces PC differentiation in SLE and suggest that a critical event might be modulation of cellular metabolism.
Collapse
Affiliation(s)
- Jingjing Ren
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Michelle D Catalina
- AMPEL BioSolutions, Charlottesville, VA, United States.,RILITE Research Institute, Charlottesville, VA, United States
| | - Kristin Eden
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Xiaofeng Liao
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Kaitlin A Read
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States.,Virginia Tech Carilion Research Institute, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Xin Luo
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Ryan P McMillan
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Matthew W Hulver
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Matthew Jarpe
- Regenacy Pharmaceuticals, Waltham, MA, United States
| | | | - Amrie C Grammer
- AMPEL BioSolutions, Charlottesville, VA, United States.,RILITE Research Institute, Charlottesville, VA, United States
| | - Peter E Lipsky
- AMPEL BioSolutions, Charlottesville, VA, United States.,RILITE Research Institute, Charlottesville, VA, United States
| | - Christopher M Reilly
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States.,Edward Via College of Osteopathic Medicine, Blacksburg, VA, United States
| |
Collapse
|
44
|
Molecular and Cellular Bases of Immunosenescence, Inflammation, and Cardiovascular Complications Mimicking "Inflammaging" in Patients with Systemic Lupus Erythematosus. Int J Mol Sci 2019; 20:ijms20163878. [PMID: 31395799 PMCID: PMC6721773 DOI: 10.3390/ijms20163878] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/04/2019] [Accepted: 08/05/2019] [Indexed: 12/19/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an archetype of systemic autoimmune disease, characterized by the presence of diverse autoantibodies and chronic inflammation. There are multiple factors involved in lupus pathogenesis, including genetic/epigenetic predisposition, sexual hormone imbalance, environmental stimulants, mental/psychological stresses, and undefined events. Recently, many authors noted that "inflammaging", consisting of immunosenescence and inflammation, is a common feature in aging people and patients with SLE. It is conceivable that chronic oxidative stresses originating from mitochondrial dysfunction, defective bioenergetics, abnormal immunometabolism, and premature telomere erosion may accelerate immune cell senescence in patients with SLE. The mitochondrial dysfunctions in SLE have been extensively investigated in recent years. The molecular basis of normoglycemic metabolic syndrome has been found to be relevant to the production of advanced glycosylated and nitrosative end products. Besides, immunosenescence, autoimmunity, endothelial cell damage, and decreased tissue regeneration could be the results of premature telomere erosion in patients with SLE. Herein, the molecular and cellular bases of inflammaging and cardiovascular complications in SLE patients will be extensively reviewed from the aspects of mitochondrial dysfunctions, abnormal bioenergetics/immunometabolism, and telomere/telomerase disequilibrium.
Collapse
|
45
|
Martirosyan A, Aminov R, Manukyan G. Environmental Triggers of Autoreactive Responses: Induction of Antiphospholipid Antibody Formation. Front Immunol 2019; 10:1609. [PMID: 31354742 PMCID: PMC6635959 DOI: 10.3389/fimmu.2019.01609] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/27/2019] [Indexed: 12/19/2022] Open
Abstract
Antiphospholipid antibodies (aPLs) comprise a diverse family of autoantibodies targeted against proteins with the affinity toward negatively charged phospholipids or protein-phospholipid complexes. Their clinical significance, including prothrombotic potential of anti-cardiolipin antibodies (aCLs), anti-β2-glycoprotein I antibodies (aβ2-GPIs), and lupus anti-coagulant (LA), is well-established. However, the ontogeny of these pathogenic aPLs remains less clear. While transient appearance of aPLs could be induced by various environmental factors, in genetically predisposed individuals these factors may eventually lead to the development of the antiphospholipid syndrome (APS). Since the first description of APS, it has been found that a wide variety of microbial and viral agents influence aPLs production and contribute to clinical manifestations of APS. Many theories attempted to explain the pathogenic potential of different environmental factors as well as a phenomenon termed molecular mimicry between β2-GPI molecule and infection-relevant structures. In this review, we summarize and critically assess the pathogenic and non-pathogenic formation of aPLs and its contribution to the development of APS.
Collapse
Affiliation(s)
- Anush Martirosyan
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Biology, Yerevan, Armenia.,Russian-Armenian (Slavonic) University, Yerevan, Armenia
| | - Rustam Aminov
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Gayane Manukyan
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Biology, Yerevan, Armenia.,Russian-Armenian (Slavonic) University, Yerevan, Armenia
| |
Collapse
|
46
|
Post-Translational Modification-Dependent Activity of Matrix Metalloproteinases. Int J Mol Sci 2019; 20:ijms20123077. [PMID: 31238509 PMCID: PMC6627178 DOI: 10.3390/ijms20123077] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/16/2019] [Accepted: 06/18/2019] [Indexed: 12/18/2022] Open
Abstract
Due to their capacity to process different proteins of the extracellular matrix (ECM), matrix metalloproteinases (MMPs) were initially described as a family of secreted proteases, functioning as main ECM regulators. However, through proteolytic processing of various biomolecules, MMPs also modulate intra- and extracellular pathways and networks. Thereby, they are functionally implicated in the regulation of multiple physiological and pathological processes. Consequently, MMP activity is tightly regulated through a combination of epigenetic, transcriptional, and post-transcriptional control of gene expression, proteolytic activation, post-translational modifications (PTMs), and extracellular inhibition. In addition, MMPs, their substrates and ECM binding partners are frequently modified by PTMs, which suggests an important role of PTMs in modulating the pleiotropic activities of these proteases. This review summarizes the recent progress towards understanding the role of PTMs (glycosylation, phosphorylation, glycosaminoglycans) on the activity of several members of the MMP family.
Collapse
|
47
|
Quantitative proteomic profiling of extracellular matrix and site-specific collagen post-translational modifications in an in vitro model of lung fibrosis. Matrix Biol Plus 2019; 1:100005. [PMID: 33543004 PMCID: PMC7852317 DOI: 10.1016/j.mbplus.2019.04.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 04/09/2019] [Accepted: 04/09/2019] [Indexed: 12/21/2022] Open
Abstract
Lung fibrosis is characterized by excessive deposition of extracellular matrix (ECM), in particular collagens, by fibroblasts in the interstitium. Transforming growth factor-β1 (TGF-β1) alters the expression of many extracellular matrix (ECM) components produced by fibroblasts, but such changes in ECM composition as well as modulation of collagen post-translational modification (PTM) levels have not been comprehensively investigated. Here, we performed mass spectrometry (MS)-based proteomics analyses to assess changes in the ECM deposited by cultured lung fibroblasts from idiopathic pulmonary fibrosis (IPF) patients upon stimulation with transforming growth factor β1 (TGF-β1). In addition to the ECM changes commonly associated with lung fibrosis, MS-based label-free quantification revealed profound effects on enzymes involved in ECM crosslinking and turnover as well as multiple positive and negative feedback mechanisms of TGF-β1 signaling. Notably, the ECM changes observed in this in vitro model correlated significantly with ECM changes observed in patient samples. Because collagens are subject to multiple PTMs with major implications in disease, we implemented a new bioinformatic platform to analyze MS data that allows for the comprehensive mapping and site-specific quantitation of collagen PTMs in crude ECM preparations. These analyses yielded a comprehensive map of prolyl and lysyl hydroxylations as well as lysyl glycosylations for 15 collagen chains. In addition, site-specific PTM analysis revealed novel sites of prolyl-3-hydroxylation and lysyl glycosylation in type I collagen. Interestingly, the results show, for the first time, that TGF-β1 can modulate prolyl-3-hydroxylation and glycosylation in a site-specific manner. Taken together, this proof of concept study not only reveals unanticipated TGF-β1 mediated regulation of collagen PTMs and other ECM components but also lays the foundation for dissecting their key roles in health and disease. The proteomic data has been deposited to the ProteomeXchange Consortium via the MassIVE partner repository with the data set identifier MSV000082958. Quantitative proteomics of TGF-β-induced changes in ECM composition and collagen PTM in pulmonary fibroblasts TGF-β promotes crosslinking and turnover as well as complex feedback mechanisms that alter fibroblast ECM homeostasis. A novel bioinformatic workflow for MS data analysis enabled global mapping and quantitation of known and novel collagen PTMs Quantitative assessment of prolyl-3-hydroxylation site occupancy and lysine-O-glycosylation microheterogeneity TGF-β1 modulates collagen PTMs in a site-specific manner that may favor collagen accumulation in lung fibrosis
Collapse
Key Words
- 3-HyP, 3-hydroxyproline
- 4-HyP, 4-hydroxyproline
- AGC, automatic gain control
- ANXA11, annexin A11
- BGN, biglycan
- COL1A1, collagen-I alpha 1 chain
- Collagen
- Collagen post-translational modifications
- DCN, decorin
- ECM, extracellular matrix
- Extracellular matrix
- FN1, fibronectin 1
- G-HyK, galactosylhydroxylysine
- GG-HyK, glucosylgalactosylhydroxylysine
- HyK, hydroxylysine
- HyP, hydroxyproline
- ILD, interstitial lung disease
- IPF, idiopathic pulmonary fibrosis
- LH, lysyl hydroxylase
- LOX(L), lysyl oxidase(-like)
- LTBP2, latent-transforming growth factor β -binding protein 2
- Lysyl glycosylation
- Lysyl hydroxylation
- P3H, prolyl-3-hydroxylase
- P4H, prolyl-4-hydroxylase
- PAI1, plasminogen activator inhibitor 1
- PCA, principal component analysis
- PLOD (LH), procollagen-lysine,2-oxoglutarate 5-dioxygenases (lysyl hydroxylases)
- PTM, post-translational modification
- Prolyl hydroxylation
- Pulmonary fibrosis
- SEMA7A, semaphorin 7a
- TGF-β, transforming growth factor β
- TGM2, transglutaminase 1
- Transforming growth factor-β
- VCAN, versican
- Xaa, Xaa position in the Gly-Xaa-Yaa repeat in triple-helical collagen
- Yaa, Yaa position in the Gly-Xaa-Yaa repeat in triple-helical collagen
- α-SMA, α-smooth muscle actin
Collapse
|
48
|
Could the high consumption of high glycaemic index carbohydrates and sugars, associated with the nutritional transition to the Western type of diet, be the common cause of the obesity epidemic and the worldwide increasing incidences of Type 1 and Type 2 diabetes? Med Hypotheses 2019; 125:41-50. [PMID: 30902150 DOI: 10.1016/j.mehy.2019.02.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 12/07/2018] [Accepted: 02/08/2019] [Indexed: 12/30/2022]
Abstract
The globally increasing incidences of Type 1 diabetes (T1DM) and Type 2 diabetes (T2DM) can have a common background. If challenged by the contemporary high level of nutritional glucose stimulation, the β-cells in genetically predisposed individuals are at risk for damage which can lead to the diseases. The fat to carbohydrate dietary shift can also contribute to the associated obesity epidemic.
Collapse
|
49
|
Abstract
High-mobility group box 1 (HMGB1) is one of the most abundant proteins in eukaryotes and the best characterized damage-associated molecular pattern (DAMP). The biological activities of HMGB1 depend on its subcellular location, context and post-translational modifications. Inside the nucleus, HMGB1 is engaged in many DNA events such as DNA repair, transcription regulation and genome stability; in the cytoplasm, its main function is to regulate the autophagic flux while in the extracellular environment, it possesses more complicated functions and it is involved in a large variety of different processes such as inflammation, migration, invasion, proliferation, differentiation and tissue regeneration. Due to this pleiotropy, the role of HMGB1 has been vastly investigated in various pathological diseases and a large number of studies have explored its function in cardiovascular pathologies. However, in this contest, the precise mechanism of action of HMGB1 and its therapeutic potential are still very controversial since is debated whether HMGB1 is involved in tissue damage or plays a role in tissue repair and regeneration. The main focus of this review is to provide an overview of the effects of HMGB1 in different ischemic heart diseases and to discuss its functions in these pathological conditions.
Collapse
|
50
|
Brandhorst TT, Klein BS. Uncertainty surrounding the mechanism and safety of the post-harvest fungicide fludioxonil. Food Chem Toxicol 2018; 123:561-565. [PMID: 30458269 DOI: 10.1016/j.fct.2018.11.037] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/09/2018] [Accepted: 11/14/2018] [Indexed: 02/06/2023]
Abstract
Fludioxonil is a phenylpyrrole pesticide that is applied to fruit and vegetable crops post-harvest to minimize losses to mold, both during transport and at point of sale. Its effectiveness is reflected in the dramatic increase in its production/usage since its introduction in 1994, an increase that has peaked in recent years as it became licenced for use abroad. Recently, doubts as to the nature of its mechanism of action have been raised. Given that the pesticide has long been known to induce stress intermediates in target and non-target organisms alike, the lack of a firmly established mechanism might be cause for concern. Troubling reports further delineate a capacity to disrupt hepatic, endocrine and neurological systems, indicating that fludioxonil may represent a health threat to consumers. In the absence of a clear, safe mechanism of action, fludioxonil should be re-evaluated for its potential to impact human health.
Collapse
Affiliation(s)
- T Tristan Brandhorst
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA.
| | - Bruce S Klein
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA; Department of Internal Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA; Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
| |
Collapse
|