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
|
Tarannum A, Arif Z, Mustafa M, Alam K, Moinuddin, Habib S. Albumin from sera of rheumatoid arthritis patients share multiple biochemical, biophysical and immunological properties with in vitro generated glyco-nitro-oxidized-albumin. J Biomol Struct Dyn 2023:1-17. [PMID: 37982266 DOI: 10.1080/07391102.2023.2283153] [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: 04/09/2023] [Accepted: 11/05/2023] [Indexed: 11/21/2023]
Abstract
The purpose of the present study is to explore the effects of endogenous stressors on structure and function of rheumatoid arthritis (RA) patients' albumin. In contrast to glycated-albumin or nitro-oxidized-albumin, high titre antibodies against glyco-nitro-oxidized-albumin were found in the sera of RA patients. Also, compared to the other two modified forms of albumin, glyco-nitro-oxidized-albumin showed highest percent inhibition. Albumin isolated from RA patients' sera displayed hyperchromicity and quenching of tyrosine and tryptophan fluorescence. Fluorescence spectroscopy studies also revealed the presence of dityrosine and advanced glycation end products in RA patient's albumin. RA patients' albumin showed weaker binding with 1-anilinonaphthalene-8-sulfonic acid dye. Secondary structure alterations were demonstrated by circular dichroism and Fourier transform infrared spectroscopy. Biochemical investigations revealed substantial decline in the availability of free side chains of amino acid residues; increased carbonyls and decreased sulfhydryls in RA patients' albumin. The functional impairment in RA patients' albumin was revealed by their low binding with bilirubin and cobalt. Liquid chromatography mass spectrometry analysis revealed the presence of Nε-(carboxymethyl) lysine and 3-nitrotyrosine in RA patients' albumin. The amyloidogenic aggregation of RA patients' albumin was confirmed by Congo red absorption and thioflavin-T fluorescence assays. The morphology of the aggregates was visualized under scanning and transmission electron microscope. From the above findings, we inferred that endogenous stress in RA patients have modified albumin and produce structural/functional abnormalities. Also, the presence of anti-glyco-nitro-oxidized-albumin antibodies along with other clinical features may be used as biomarker for the diagnosis and assessment of treatment responses in RA patients.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Akhlas Tarannum
- Department of Biochemistry, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Zarina Arif
- Department of Biochemistry, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Mohd Mustafa
- Department of Biochemistry, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Khursheed Alam
- Department of Biochemistry, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Moinuddin
- Department of Biochemistry, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Safia Habib
- Department of Biochemistry, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| |
Collapse
|
3
|
Ermurat S, Güler Kazanci E, Toka DI, Ayar K, Eren FK, Neşelioğlu S. Evaluation of thiol/disulfide hemostasis and serum Ischemia modified albumin as oxidative stress biomarkers in systemic lupus erythematosus patients: Relationship with major organ involvement and disease activity. Lupus 2022; 31:1355-1366. [PMID: 35850631 DOI: 10.1177/09612033221115628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVES We aimed to compare thiol/disulfide hemostasis and serum ischemia-modified albumin (IMA) levels, which are indicators of oxidative stress (OS), in patients with systemic lupus erythematosus (SLE), with the healthy control (HC) group and to evaluate the relationship of these parameters with disease activity and major organ involvement. MATERIAL-METHODS Eighty-four SLE patients and 96 HCs were included in this study. The disease activity of SLE patients was calculated using The Systemic Lupus Erythematosus Disease Activity Index 2000 (SLEDAI-2K). Patients with SLEDAI-2K ≤ 5 were classified as low disease activity (LDA) and those with SLEDAI-2K > 6 as high disease activity (HDA). Thiol/disulfide hemostasis was evaluated using a new automated method and natural thiol (NT), total thiol (TT), disulfide (SS) levels, SS/NT, SS/TT, NT/TT ratios, and serum IMA levels were recorded. RESULTS NT and TT levels were significantly lower (490.11 ± 123.61 vs 536.96 ± 86.05, p = 0.003) (532.56 ± 125.80 vs 565.72 ± 89.82, p = 0.046), SS level (21.22 ± 11.75 vs 13.37 ± 9.31, p < 0.001) was higher, and SS/TT (4.64 ± 2.93 vs 2.52 ± 1.82, p < 0.001) and SS/NT (4.12 ± 2.33 vs 2.35 ± 1.59, p < 0.001) ratios were significantly higher in SLE patients compared to HCs. IMA values were not different between the two groups (p = 0.920). NT (449.84 ± 136.98 vs 520.32 ± 104.11, p = 0.012) and TT levels (492.01 ± 138.45±562.97 ± 107.09, p = 0.013) were significantly lower and serum IMA levels (0.802 ± 0.089 vs 0.764 ± 0.040, p = 0.023) were significantly higher in SLE patients with HDA than in LDA patients. There was a weak negative correlation between NT (r = -0.284, p=0.009) (r = -0.291, p = 0.007) and TT levels (r = -0.281, p = 0.010) (r = -0.289, p = 0.008) and a weak positive correlation between IMA levels (r = 0.279, p = 0.011) (r = 0.263, p = 0.016) and SLEDAI-2K, and major organ involvement. CONCLUSION It is thought that thiol/disulfide hemostasis and IMA levels may be used as ideal biomarkers of OS in SLE patients and may reflect the disease activity and major organ involvement.
Collapse
Affiliation(s)
- Selime Ermurat
- Pediatric Hematology, 147003Bursa Yüksek İhtisas Training and Research Hospital, Bursa, Turkey
| | - Elif Güler Kazanci
- Pediatric Hematology, 147003Bursa Yüksek İhtisas Training and Research Hospital, Bursa, Turkey
| | - Dilara Isiyel Toka
- Internal Medicine, 147003Bursa Yüksek İhtisas Training and Research Hospital, Bursa, Turkey
| | - Koray Ayar
- Pediatric Hematology, 147003Bursa Yüksek İhtisas Training and Research Hospital, Bursa, Turkey
| | | | - Salim Neşelioğlu
- Rheumatology, 147003Bursa Yüksek İhtisas Training and Research Hospital, Bursa, Turkey
| |
Collapse
|
4
|
Miglioranza Scavuzzi B, Holoshitz J. Endoplasmic Reticulum Stress, Oxidative Stress, and Rheumatic Diseases. Antioxidants (Basel) 2022; 11:1306. [PMID: 35883795 PMCID: PMC9312221 DOI: 10.3390/antiox11071306] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 12/10/2022] Open
Abstract
BACKGROUND The endoplasmic reticulum (ER) is a multi-functional organelle responsible for cellular homeostasis, protein synthesis, folding and secretion. It has been increasingly recognized that the loss of ER homeostasis plays a central role in the development of autoimmune inflammatory disorders, such as rheumatic diseases. Purpose/Main contents: Here, we review current knowledge of the contribution of ER stress to the pathogenesis of rheumatic diseases, with a focus on rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). We also review the interplay between protein folding and formation of reactive oxygen species (ROS), where ER stress induces oxidative stress (OS), which further aggravates the accumulation of misfolded proteins and oxidation, in a vicious cycle. Intervention studies targeting ER stress and oxidative stress in the context of rheumatic diseases are also reviewed. CONCLUSIONS Loss of ER homeostasis is a significant factor in the pathogeneses of RA and SLE. Targeting ER stress, unfolded protein response (UPR) pathways and oxidative stress in these diseases both in vitro and in animal models have shown promising results and deserve further investigation.
Collapse
Affiliation(s)
| | - Joseph Holoshitz
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA;
| |
Collapse
|
5
|
Moschetti L, Piantoni S, Vizzardi E, Sciatti E, Riccardi M, Franceschini F, Cavazzana I. Endothelial Dysfunction in Systemic Lupus Erythematosus and Systemic Sclerosis: A Common Trigger for Different Microvascular Diseases. Front Med (Lausanne) 2022; 9:849086. [PMID: 35462989 PMCID: PMC9023861 DOI: 10.3389/fmed.2022.849086] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/04/2022] [Indexed: 11/13/2022] Open
Abstract
This review describes the complex interplay between inflammation, vasculopathy and fibrosis that involve the heart and peripheral small vessels, leading to endothelial stiffness, vascular damage, and early aging in patients with systemic lupus erythematosus and systemic sclerosis, which represents two different models of vascular dysfunction among systemic autoimmune diseases. In fact, despite the fact that diagnostic methods and therapies have been significantly improved in the last years, affected patients show an excess of cardiovascular mortality if compared with the general population. In addition, we provide a complete overview on the new techniques which are used for the evaluation of endothelial dysfunction in a preclinical phase, which could represent a new approach in the assessment of cardiovascular risk in these patients.
Collapse
Affiliation(s)
- Liala Moschetti
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Silvia Piantoni
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- *Correspondence: Silvia Piantoni,
| | - Enrico Vizzardi
- Cardiology Unit, ASST Spedali Civili of Brescia, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | | | - Mauro Riccardi
- Cardiology Unit, ASST Spedali Civili of Brescia, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Franco Franceschini
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Ilaria Cavazzana
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
6
|
Obiweluozo PE, Onwurah CN, Uzodinma UE, Dike IC, Onwurah AI. Particulate air-borne pollutants in Port Harcourt could contaminate recreational pools; toxicity evaluation and children's health risk assessment. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:2342-2352. [PMID: 34370197 DOI: 10.1007/s11356-021-15704-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 07/24/2021] [Indexed: 06/13/2023]
Abstract
Air pollution in Port Harcourt metropolis exacerbated by ambient air-borne black soot particles in the past 4 years has become a great concern especially for children who indulge in recreational pools' activities. This study was therefore carried out to evaluate the toxicities of recreational pools in Port Harcourt City and hence perform preliminary health risk assessment. Five recreational pools (4 outdoor and 1 indoor) were used out of the randomly selected 8, from 30 that were identified. Toxicity assessment was carried out using "Ostracods-linked mathematical model" while risk assessment was by analysing protein oxidation and hepatotoxicity in the hepatocytes of exposed rats. Questionnaire-based approach was used to elicit relevant information from children (10-13 years) who consistently engaged in recreational pool activities. The pH and temperature and bacteriological examination of the pools were also carried out. Results show that all the pools were in fairly good sanitary conditions based on the highest mean bacterial counts (2.33±0.57/100 ml), while all were slightly acidic and with normal temperature range. The toxicity indices of outdoor pools (SP 1) and (SP 5) are respectively 20.8 and 49.0 units, while that of outdoor pools (SP 2) and (SP 3) are the same (42.5 units). "No-observed-effect-toxicity" (NOET) was observed for the indoor pool (SP 4). Analysis of the structured questionnaires inferred that none of the children traced has or has had any of the recreational water illnesses. However, results of protein oxidation in rats and hepatotoxicity of rats' hepatocytes suggest that the exposed children may be at oxidative stress-related risks in future especially if the children continue, without adequate precaution, in the use of these pools. It is recommended that further monitoring of these children be continued while measures such as frequent changes of the pool water are ensured.
Collapse
Affiliation(s)
- Patience E Obiweluozo
- Childhood and Environmental Education Research Group, Department of Educational Foundations, University of Nigeria, Nsukka, Nigeria
| | - Chimezie N Onwurah
- Childhood and Environmental Education Research Group, Department of Educational Foundations, University of Nigeria, Nsukka, Nigeria
| | - Uche E Uzodinma
- Childhood and Environmental Education Research Group, Department of Educational Foundations, University of Nigeria, Nsukka, Nigeria
| | - Ibiwari C Dike
- Childhood and Environmental Education Research Group, Department of Educational Foundations, University of Nigeria, Nsukka, Nigeria
| | - Arinze I Onwurah
- Centre for Environmental Management and Control (CEMAC), University of Nigeria, Enugu Campus, Nsukka, Nigeria.
| |
Collapse
|
7
|
Nocella C, Bartimoccia S, Cammisotto V, D’Amico A, Pastori D, Frati G, Sciarretta S, Rosa P, Felici C, Riggio O, Calogero A, Carnevale R. Oxidative Stress in the Pathogenesis of Antiphospholipid Syndrome: Implications for the Atherothrombotic Process. Antioxidants (Basel) 2021; 10:antiox10111790. [PMID: 34829661 PMCID: PMC8615138 DOI: 10.3390/antiox10111790] [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: 09/29/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 12/05/2022] Open
Abstract
Atherothrombosis is a frequent complication of the clinical history of patients with antiphospholipid syndrome (APS). Both atherothrombosis and APS are characterized by increased oxidative stress. Oxidative modifications are implicated in the formation of antiphospholipid antibodies, which in turn may favour the oxidative imbalance by increasing the production of reactive oxidant species (ROS) or by a direct interaction with pro-oxidant/antioxidant enzymes. As a result of these processes, APS patients suffer from an oxidative imbalance that may contribute to the progression of the atherosclerotic process and to the onset of ischemic thrombotic complications. The aim of this review is to describe mechanisms implicated in the formation of ROS in APS patients and their involvement in the atherothrombotic process. We also provide an overview of potential therapeutic approaches to blunt oxidative stress and to prevent atherothrombotic complications in these patients.
Collapse
Affiliation(s)
- Cristina Nocella
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (C.N.); (D.P.)
| | - Simona Bartimoccia
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (G.F.); (S.S.); (P.R.); (C.F.); (A.C.)
| | - Vittoria Cammisotto
- Department of General Surgery and Surgical Specialty Paride Stefanini, Sapienza University of Rome, 00161 Rome, Italy;
| | - Alessandra D’Amico
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy;
| | - Daniele Pastori
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00161 Rome, Italy; (C.N.); (D.P.)
| | - Giacomo Frati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (G.F.); (S.S.); (P.R.); (C.F.); (A.C.)
- Department of AngioCardioNeurology, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Sebastiano Sciarretta
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (G.F.); (S.S.); (P.R.); (C.F.); (A.C.)
- Department of AngioCardioNeurology, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Paolo Rosa
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (G.F.); (S.S.); (P.R.); (C.F.); (A.C.)
| | - Chiara Felici
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (G.F.); (S.S.); (P.R.); (C.F.); (A.C.)
| | - Oliviero Riggio
- Department of Translational and Precision Medicine, “Sapienza” University of Rome, 00161 Rome, Italy;
- Faculty of Medicine and Surgery, Course E, Sapienza University of Rome, 04100 Latina, Italy;
| | - Antonella Calogero
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (G.F.); (S.S.); (P.R.); (C.F.); (A.C.)
| | - Roberto Carnevale
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy; (S.B.); (G.F.); (S.S.); (P.R.); (C.F.); (A.C.)
- Mediterranea, Cardiocentro, 80122 Napoli, Italy
- Correspondence: ; Tel./Fax: +39-0773-175-7245
| | - SMiLe Group
- Faculty of Medicine and Surgery, Course E, Sapienza University of Rome, 04100 Latina, Italy;
| |
Collapse
|
8
|
Cellular Response against Oxidative Stress, a Novel Insight into Lupus Nephritis Pathogenesis. J Pers Med 2021; 11:jpm11080693. [PMID: 34442337 PMCID: PMC8401250 DOI: 10.3390/jpm11080693] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023] Open
Abstract
The interaction of reactive oxygen species (ROS) with lipids, proteins, nucleic acids and hydrocarbonates promotes acute and chronic tissue damage, mediates immunomodulation and triggers autoimmunity in systemic lupus erythematous (SLE) patients. The aim of the study was to determine the pathophysiological mechanisms of the oxidative stress-related damage and molecular mechanisms to counteract oxidative stimuli in lupus nephritis. Our study included 38 SLE patients with lupus nephritis (LN group), 44 SLE patients without renal impairment (non-LN group) and 40 healthy volunteers as control group. In the present paper, we evaluated serum lipid peroxidation, DNA oxidation, oxidized proteins, carbohydrate oxidation, and endogenous protective systems. We detected defective DNA repair mechanisms via 8-oxoguanine-DNA-glycosylase (OGG1), the reduced regulatory effect of soluble receptor for advanced glycation end products (sRAGE) in the activation of AGE-RAGE axis, low levels of thiols, disulphide bonds formation and high nitrotyrosination in lupus nephritis. All these data help us to identify more molecular mechanisms to counteract oxidative stress in LN that could permit a more precise assessment of disease prognosis, as well as developing new therapeutic targets.
Collapse
|
9
|
Cytokine Profiling in Chinese SLE Patients: Correlations with Renal Dysfunction. J Immunol Res 2021; 2020:8146502. [PMID: 33134397 PMCID: PMC7568803 DOI: 10.1155/2020/8146502] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 12/19/2022] Open
Abstract
Background Systemic lupus erythematosus (SLE) is a chronic, systemic autoimmune disease that commonly causes kidney damage. Therefore, we measured plasma levels of cytokines that may be related to renal dysfunction in SLE patients. Methods To explore the differences between SLE patients with renal dysfunction and healthy volunteers, the levels of cytokines in plasma were screened using a human cytokine antibody array. Then, we chose fourteen of the elevated cytokines for verification with an expanded sample size by a human magnetic Luminex assay. Plasma samples were isolated from SLE patients (n = 72) and healthy volunteers (n = 8). Results Cytokine antibody array data showed elevated plasma cytokines in SLE patients with renal dysfunction compared with healthy volunteers. By using the human magnetic Luminex assay, we found that plasma levels of CHI3L1, GDF-15, IGFBP-2, MIF, ST2, TFF3, and uPAR were significantly higher in SLE patients than in healthy volunteers. Plasma levels of CXCL4 were significantly lower in the active group than in the inactive group, and plasma levels of CHI3L1, IGFBP-2, MIF, and MPO were significantly higher in the active group than in the inactive group. We also analyzed the correlation between plasma cytokine levels and the SLEDAI-2K, and our results showed that the plasma levels of the fourteen selected cytokines were weakly correlated or not correlated with the SLEDAI-2K. We further analyzed the correlation between cytokines and renal dysfunction. Plasma levels of GDF-15 and TFF3 were highly positively correlated with serum creatinine levels and 24-hour urine protein levels. Conclusion Our data suggest that plasma levels of GDF-15 and TFF3 are potential renal dysfunction markers in SLE patients, but plasma levels of these cytokines are not correlated with the SLEDAI-2K. Further study is warranted to determine how these cytokines regulate inflammatory responses and renal dysfunction in SLE.
Collapse
|
10
|
Banerjee N, Wang H, Wang G, Boor PJ, Khan MF. Redox-sensitive Nrf2 and MAPK signaling pathways contribute to trichloroethene-mediated autoimmune disease progression. Toxicology 2021; 457:152804. [PMID: 33930529 PMCID: PMC8230612 DOI: 10.1016/j.tox.2021.152804] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 12/15/2022]
Abstract
Trichloroethene (TCE) exposure is associated with the induction of autoimmune diseases (ADs). Although oxidative stress plays a major role in TCE-mediated autoimmunity, the underlying molecular mechanisms still need to be delineated. Dysregulation of redox-sensitive nuclear factor (erythroid-derived 2)-like2 (Nrf2), resulting in uncontrolled antioxidant and cytoprotective genes, and pro-inflammatory MAPK signaling pathways could be critical in TCE-mediated disease progression. This study was, therefore, focused on establishing status and contribution of Nrf2 and MAPK signaling in TCE-mediated inflammatory and autoimmune responses, especially during disease progression. To achieve these objectives, time-response studies were conducted by treating female MRL+/+ mice with TCE (0.5 mg/mL, a dose relevant to human exposure) for 24, 36 and 52 wks. TCE exposure led to reduction in Nrf2 expression, but increased phos-NF-κB (p65) and iNOS along with increased phosphorylation of MAPKs (p38, ERK and JNK) and downstream pro-inflammatory cytokines IL-12, TNF-α and RANTES in the livers in a time-dependent manner. These changes were also associated with time-dependent increases in liver protein carbonyls and induction of serum anti-dsDNA antibodies (marker of systemic lupus erythematosus disease), further supporting the role of oxidative stress and Nrf2/MAPK signaling in TCE-mediated autoimmune response progression. The mechanistic role of MAPK in TCE-mediated autoimmunity was further established by treating MRL+/+ mice with sulforaphane (SFN; 8 mg/kg, i.p., every other day) along with TCE (10 mmol/kg, i.p., every 4th day) for 6 wks using an established protocol, and by in vitro treatment of T cells with dichloroacetyl chloride (a TCE metabolite) with/without p38 MAPK inhibitor. SFN treatment attenuated the TCE-mediated phosphorylation of p38 MAPK. More importantly, treatment with SFN or p38 inhibitor led to suppression of downstream pro-inflammatory cytokines IL-12 and TNF-α. These findings thus support the contribution of Nrf2 and MAPK signaling pathways and help in delineating novel potential therapeutic targets against TCE-mediated autoimmunity.
Collapse
Affiliation(s)
- Nivedita Banerjee
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, United States
| | - Hui Wang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, United States
| | - Gangduo Wang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, United States
| | - Paul J Boor
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, United States
| | - M Firoze Khan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, United States.
| |
Collapse
|
11
|
de Oliveira MAA, Mallmann NH, de Souza GKBB, de Jesus Bacha T, Lima ES, de Lima DSN, de Souza Passos LF, de Souza Gonçalves M, de Moura Neto JP. Glutathione S-transferase, catalase, and mitochondrial superoxide dismutase gene polymorphisms modulate redox potential in systemic lupus erythematosus patients from Manaus, Amazonas, Brazil. Clin Rheumatol 2021; 40:3639-3649. [PMID: 33745084 DOI: 10.1007/s10067-021-05680-0] [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: 07/22/2020] [Revised: 02/28/2021] [Accepted: 03/02/2021] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To investigate the frequency of glutathione S-transferase (GST), catalase, and SOD2 genetic polymorphisms and their correlation with SLE. METHODS A total of 290 females (patients = 151; controls= 139) were recruited. Multiplex PCR was performed for genotyping GSTM1 and GSTT1 genes, whereas real-time qPCR was used for determination of SNPs: CAT C262T, SOD2 C47T, GSTP1 A313G and GSTP1 IVS6 -C16T. RESULTS Thiol levels are decreased in SLE patients (p<0.001), while MDA levels were significantly higher (p<0.001) and those carrying the polymorphisms had higher rates of oxidative stress. Patients with double null deletion GSTT1null/GSTM1null had a frequency almost five times higher than the controls (p<0.001, OR 4.81, CI 1.98-12.11). SLE patients had a lower wild-type frequency of SOD2CC allele compared to controls (12.4% vs 27.3%). Statistical significances were observed on the association between the GSTT1null and GSTM1null with SOD2mut (p<0.001, OR 0.15, CI 0.05-0.47), with GSTP1 A303G (p=0.012, OR 0.19, CI 0.05-0.69), and with GSTP1 IVS6 (p=0.008, OR 0.14, CI 0.03-0.63). The same was observed between SOD2 C47T with GSTP1 A303G (p=0.09, OR 0.27, CI 0.09-0.74) and GSTP1 IVS6 (p=0.036, OR 0.41, CI 0.18-0.92). CONCLUSIONS The deletion GSTT1null/GSTM1null may contribute to the increased of the oxidative stress in SLE patients. Isolated GSTP1 and CAT polymorphisms do not seem to influence the increased oxidative stress, neither SLE clinical manifestations. SOD2 47CT/TT allele may have greater oxidative stress due to structural change in the protein and decreased H2O2 production. The combination of polymorphic genes may be involved in the pathogenesis of the disease. Key points • Major question of our paper: Many studies have shown that the antioxidant status levels are decreased in patients with SLE, especially in severe stages of disease. We believe that this paper will be of interest to the readership of your journal had the involvement of polymorphisms and mutations in several genes that contribute to the genetic etiology of SLE, suggesting that these may influence the mechanisms of disease. • Our results. Thiol level was significantly (p<.001) lower and MDA level significantly increased (p<.001) among SLE patients. Those carrying the polymorphisms had higher rates of oxidative stress. SLE Patients had a frequency almost five times higher of double null deletion GSTT1null/GSTM1null than the controls. SLE Patients had a lower wild type frequency of SOD2CC allele compared to controls (12.4% vs 27.3%). We believed the deletion GSTT1null/GSTM1null may contribute to the increased of the oxidative stress in SLE patients while carriers of the mutant SOD2 47CT/TT allele may have greater oxidative stress due to structural change in the protein and decreased H2O2 production. The combination of polymorphic genes may be involved in the pathogenesis of the disease. • Implications of our results: Evidence for the involvement of genetic factors in severe clinical to lupus is compelling. This manuscript shows genetic insights in pathogenic pathways that may lead to severe clinical implications to LES. Therefore, it is necessary to understand their impact on overall disease pathogenesis and prognosis in these patients. We understand from general consensus about environmental factors can modify disease, however, maybe just in individuals who have a permissive genetic background. Even that no single gene predisposes some individuals to LES, we believe the genetic factors described in this manuscript are important elements in susceptibility to severe clinical to LES.
Collapse
Affiliation(s)
- Marco Aurélio Almeida de Oliveira
- Faculdade de Ciências Farmacêuticas, Universidade Federal do Amazonas, Avenida General Rodrigo Otávio Jordão Ramos 6200 - Coroado, Manaus - AM, CEP: 69067-005, Brazil
| | - Neila Hiraishi Mallmann
- Faculdade de Ciências Farmacêuticas, Universidade Federal do Amazonas, Avenida General Rodrigo Otávio Jordão Ramos 6200 - Coroado, Manaus - AM, CEP: 69067-005, Brazil
| | - Giselle Katiane Bonfim Bacellar de Souza
- Faculdade de Ciências Farmacêuticas, Universidade Federal do Amazonas, Avenida General Rodrigo Otávio Jordão Ramos 6200 - Coroado, Manaus - AM, CEP: 69067-005, Brazil
| | - Thiago de Jesus Bacha
- Faculdade de Ciências Farmacêuticas, Universidade Federal do Amazonas, Avenida General Rodrigo Otávio Jordão Ramos 6200 - Coroado, Manaus - AM, CEP: 69067-005, Brazil
| | - Emerson Silva Lima
- Faculdade de Ciências Farmacêuticas, Universidade Federal do Amazonas, Avenida General Rodrigo Otávio Jordão Ramos 6200 - Coroado, Manaus - AM, CEP: 69067-005, Brazil
| | | | | | | | | |
Collapse
|
12
|
Onozuka D, Nakamura Y, Tsuji G, Furue M. Mortality in Yusho patients exposed to polychlorinated biphenyls and polychlorinated dibenzofurans: a 50-year retrospective cohort study. Environ Health 2020; 19:119. [PMID: 33228703 PMCID: PMC7685647 DOI: 10.1186/s12940-020-00680-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 11/17/2020] [Indexed: 05/03/2023]
Abstract
BACKGROUND In 1968, the Yusho incident resulted in accidental exposure to polychlorinated biphenyls (PCBs), polychlorinated dibenzofurans (PCDFs), and related compounds in Japan. This study updated the risk of mortality in Yusho patients. METHODS We obtained updated cohort data for all Yusho patients for the period 1968-2017. We calculated standardized mortality ratios (SMRs) for all-cause and cause-specific mortality over a 50-year follow-up period compared with the general population in Japan. RESULTS A total of 1664 Yusho patients with 63,566 person-years of follow up were included in the analysis. Among males, excess mortality was observed for all cancers (SMR: 1.22, 95% confidence interval [CI]: 1.02 to 1.45) and lung cancer (SMR: 1.59, 95% CI: 1.12 to 2.19). Among females, increased mortality was observed for liver cancer (SMR: 2.05, 95% CI: 1.02 to 3.67). No significant increase was seen in non-cancer-related mortality compared with the general population. CONCLUSIONS Carcinogenic risk in humans after exposure to PCBs and PCDFs remains higher among Yusho patients. Our findings suggest the importance of care engagement and optimum management to deal with the burden of Yusho disease.
Collapse
Affiliation(s)
- Daisuke Onozuka
- Department of Preventive Medicine and Epidemiology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibeshin-machi, Suita, Osaka, 564-8565 Japan
- Department of Health Care Administration and Management, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuko Nakamura
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, Fukuoka, Japan
| | - Gaku Tsuji
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, Fukuoka, Japan
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masutaka Furue
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, Fukuoka, Japan
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
13
|
Tarannum A, Arif Z, Alam K, Moinuddin. Glycation, nitro-oxidation and glyco-nitro-oxidation of human serum albumin: A physico-chemical study. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.127991] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
14
|
Postprandial Glycemia, Insulinemia, and Antioxidant Status in Healthy Subjects after Ingestion of Bread made from Anthocyanin-Rich Riceberry Rice. Nutrients 2020; 12:nu12030782. [PMID: 32188005 PMCID: PMC7146297 DOI: 10.3390/nu12030782] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/13/2020] [Accepted: 03/13/2020] [Indexed: 02/07/2023] Open
Abstract
Riceberry rice, a gluten-free grain, contains many nutrient components, including carbohydrates, proteins, certain fatty acids, and micronutrients, as well as bioactive non-nutrient compounds, such as polyphenolic compounds. This study aimed to evaluate the effect of bread made from anthocyanin-rich Riceberry rice on the postprandial glycemic response, glucagon-like peptide-1 (GLP-1), antioxidant status, and subjective ratings of appetite. In the crossover design, 16 healthy participants (six men and 10 women) completed four sessions involving blood collection in the fasting state and at 30, 60, 90, 120, 150, and 180 min after food consumption (50 g of available carbohydrate) in a randomized order: 1) glucose solution, 2) wheat bread (WB), 3) Riceberry rice bread (RRB), and 4) Hom Mali bread (HMB). Consumption of RRB resulted in significantly lower postprandial plasma glucose concentration at 30 and 60 min when compared to HMB. No difference in postprandial glucose concentration between RRB and WB was observed. In addition, postprandial plasma insulin showed a significant decrease in the group which received RRB at 15 and 60 min, as compared to HMB. In comparison with 50 g of glucose, as a reference, the glycemic index (GI) of RRB, WB, and HMB was 69.3 ± 4.4, 77.8 ± 4.6, and 130.6 ± 7.9, respectively. Interestingly, the ferric-reducing ability of plasma (FRAP) level was shown to significantly increase after consumption of RRB. In the meantime, a significant decrease in the postprandial FRAP level was also observed following an intake of WB and HMB. All breads caused increases in the postprandial plasma protein thiol group and had similar effects on hunger, fullness, desire to eat, and satiety ratings. However, consumption of RBB, WB, and HMB did not change plasma GLP-1 and malondialdehyde (MDA) levels when compared to the baseline. The findings suggest that anthocyanin-rich Riceberry rice can be a natural ingredient for gluten-free bread which reduced glycemic response together with improvement of antioxidant status in healthy subjects.
Collapse
|
15
|
Sciatti E, Cavazzana I, Vizzardi E, Bonadei I, Fredi M, Taraborelli M, Ferizi R, Metra M, Tincani A, Franceschini F. Systemic Lupus Erythematosus and Endothelial Dysfunction: A Close Relationship. Curr Rheumatol Rev 2020; 15:177-188. [PMID: 30474532 DOI: 10.2174/1573397115666181126105318] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/04/2018] [Accepted: 11/14/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Accelerated atherosclerosis, responsible for premature cardiovascular disease, has been estimated to develop or progress in 10% of systemic lupus erythematosus (SLE) patients each year and to be 6-fold more frequent in SLE compared with the general population. The mechanisms underlying accelerated atherosclerosis in SLE are complex and involve classical and "non-classical" cardiovascular risk factors. Subclinical and disseminated atherosclerosis is associated with endothelial dysfunction and arterial stiffness. OBJECTIVE The aim of this review is to analyze the association between SLE and endothelial dysfunction. RESULTS AND CONCLUSION Different mechanisms have been proposed to explain the prevalence of endothelial dysfunction in SLE, which are briefly reported in this review: impaired clearance of apoptotic cells, oxidative stress markers, B cell activation with different circulating autoantibodies, different subtypes of T lymphocytes, cytokine cascade. Several studies and meta-analyses show a significant trend towards a prevalence of subclinical accelerated atherosclerosis in patients with SLE compared with healthy controls, since childhood. Based on general considerations, we suggest a multidisciplinary management to assess endothelial dysfunction at the diagnosis of the disease and to periodically search for and treat the traditional cardiovascular risk factors. Prospective studies are needed to confirm the benefits of this management.
Collapse
Affiliation(s)
- Edoardo Sciatti
- Cardiology Unit, University and ASST Spedali Civili, Brescia, Italy
| | - Ilaria Cavazzana
- Rheumatology and Clinical Immunolgy Unit, University and ASST Spedali Civili, Brescia, Italy
| | - Enrico Vizzardi
- Cardiology Unit, University and ASST Spedali Civili, Brescia, Italy
| | - Ivano Bonadei
- Cardiology Unit, University and ASST Spedali Civili, Brescia, Italy
| | - Micaela Fredi
- Rheumatology and Clinical Immunolgy Unit, University and ASST Spedali Civili, Brescia, Italy
| | - Mara Taraborelli
- Internal Medicine Unit, ASST Franciacorta, Chiari, Brescia, Italy
| | - Romina Ferizi
- Cardiology Unit, University and ASST Spedali Civili, Brescia, Italy
| | - Marco Metra
- Cardiology Unit, University and ASST Spedali Civili, Brescia, Italy
| | - Angela Tincani
- Rheumatology and Clinical Immunolgy Unit, University and ASST Spedali Civili, Brescia, Italy
| | - Franco Franceschini
- Rheumatology and Clinical Immunolgy Unit, University and ASST Spedali Civili, Brescia, Italy
| |
Collapse
|
16
|
Jeremic I, Djuric O, Nikolic M, Vlajnic M, Nikolic A, Radojkovic D, Bonaci-Nikolic B. Neutrophil extracellular traps-associated markers are elevated in patients with systemic lupus erythematosus. Rheumatol Int 2019; 39:1849-1857. [PMID: 31444555 DOI: 10.1007/s00296-019-04426-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 08/12/2019] [Indexed: 01/21/2023]
Abstract
Neutrophil extracellular traps (NETs) are the main source of autoantigens in systemic lupus erythematosus (SLE). The aim of this study was to evaluate the clinical importance of NETs-associated markers in SLE. We compared NETs-associated markers in SLE patients (n = 111) with healthy controls (n = 50). Moreover, in 35 patients with drug-naïve SLE (n = 35), we investigated correlation between NETs-associated markers [DNase I concentration, myeloperoxidase (MPO) activity, anti-MPO antibodies, cell-free DNA (cfDNA), NETolytic activity] with serological parameters [anti-dsDNA antibodies, C3, C4 and B-cell activating factor (BAFF) levels] and disease activity measured by modified SLE Disease Activity Index (M-SLEDAI-2K). In comparison with healthy controls, SLE patients had higher cfDNA, MPO activity, anti-MPO antibodies (p < 0.001), BAFF and DNase I concentration (p < 0.01). Contrary, NETolytic activity was lower in SLE patients (p < 0.05), despite higher concentration of DNase I. MPO activity and cfDNA levels showed correlation with DNase I concentration (p < 0.001, p < 0.01, respectively). BAFF levels correlated with cfDNA, DNase I concentration and MPO activity (p < 0.05). Anti-dsDNA antibodies showed correlation with MPO activity (p < 0.01), cfDNA and BAFF levels (p < 0.001). Anti-dsDNA and C3 levels were independent predictors of M-SLEDAI-2K in multivariate analysis (p < 0.01). We demonstrated that sera of SLE patients have decreased NETolytic activity, leading to increased levels of various NETs-associated markers, which correlate with anti-dsDNA antibodies in drug-naïve SLE. We showed that BAFF participates in a complex relationship between NETosis and anti-dsDNA antibodies production. These findings have important implications for a better understanding of SLE pathogenesis and development of therapy that inhibits NETs persistence and disease progression.
Collapse
Affiliation(s)
- Ivica Jeremic
- School of Medicine, Institute of Rheumatology, University of Belgrade, Resavska 69, Belgrade, 11000, Serbia.
| | - Olivera Djuric
- School of Medicine, Institute of Epidemiology, University of Belgrade, Belgrade, Serbia
| | - Milos Nikolic
- School of Medicine, Clinic of Dermatovenereology, Clinical Centre of Serbia, University of Belgrade, Belgrade, Serbia
| | - Marina Vlajnic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Nikolic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Dragica Radojkovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Branka Bonaci-Nikolic
- School of Medicine, Clinic of Allergy and Immunology, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
17
|
Dey-Rao R, Sinha AA. In silico Analyses of Skin and Peripheral Blood Transcriptional Data in Cutaneous Lupus Reveals CCR2-A Novel Potential Therapeutic Target. Front Immunol 2019; 10:640. [PMID: 30984198 PMCID: PMC6450170 DOI: 10.3389/fimmu.2019.00640] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 03/08/2019] [Indexed: 12/17/2022] Open
Abstract
Cutaneous lesions feature prominently in lupus erythematosus (LE). Yet lupus and its cutaneous manifestations exhibit extraordinary clinical heterogeneity, making it imperative to stratify patients with varying organ involvement based on molecular criteria that may be of clinical value. We conducted several in silico bioinformatics-based analyses integrating chronic cutaneous lupus erythematosus (CCLE)-skin and blood expression profiles to provide novel insights into disease mechanisms and potential future therapy. In addition to substantiating well-known prominent apoptosis and interferon related response in both tissue environments, the overrepresentation of GO categories in the datasets, in the context of existing literature, led us to model a “disease road-map” demonstrating a coordinated orchestration of the autoimmune response in CCLE reflected in three phases: (1) initiation, (2) amplification, and (3) target damage in skin. Within this framework, we undertook in silico interactome analyses to identify significantly “over-connected” genes that are potential key functional players in the metabolic reprogramming associated with skin pathology in CCLE. Furthermore, overlapping and distinct transcriptional “hot spots” within CCLE skin and blood expression profiles mapping to specified chromosomal locations offer selected targets for identifying disease-risk genes. Lastly, we used a novel in silico approach to prioritize the receptor protein CCR2, whose expression level in CCLE tissues was validated by qPCR analysis, and suggest it as a drug target for use in future potential CCLE therapy.
Collapse
Affiliation(s)
- Rama Dey-Rao
- Department of Dermatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Animesh A Sinha
- Department of Dermatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
18
|
Smallwood MJ, Nissim A, Knight AR, Whiteman M, Haigh R, Winyard PG. Oxidative stress in autoimmune rheumatic diseases. Free Radic Biol Med 2018; 125:3-14. [PMID: 29859343 DOI: 10.1016/j.freeradbiomed.2018.05.086] [Citation(s) in RCA: 204] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 05/15/2018] [Accepted: 05/28/2018] [Indexed: 12/23/2022]
Abstract
The management of patients with autoimmune rheumatic diseases such as rheumatoid arthritis (RA) remains a significant challenge. Often the rheumatologist is restricted to treating and relieving the symptoms and consequences and not the underlying cause of the disease. Oxidative stress occurs in many autoimmune diseases, along with the excess production of reactive oxygen species (ROS) and reactive nitrogen species (RNS). The sources of such reactive species include NADPH oxidases (NOXs), the mitochondrial electron transport chain, nitric oxide synthases, nitrite reductases, and the hydrogen sulfide producing enzymes cystathionine-β synthase and cystathionine-γ lyase. Superoxide undergoes a dismutation reaction to generate hydrogen peroxide which, in the presence of transition metal ions (e.g. ferrous ions), forms the hydroxyl radical. The enzyme myeloperoxidase, present in inflammatory cells, produces hypochlorous acid, and in healthy individuals ROS and RNS production by phagocytic cells is important in microbial killing. Both low molecular weight antioxidant molecules and antioxidant enzymes, such as superoxide dismutase, catalase, glutathione peroxidase, and peroxiredoxin remove ROS. However, when ROS production exceeds the antioxidant protection, oxidative stress occurs. Oxidative post-translational modifications of proteins then occur. Sometimes protein modifications may give rise to neoepitopes that are recognized by the immune system as 'non-self' and result in the formation of autoantibodies. The detection of autoantibodies against specific antigens, might improve both early diagnosis and monitoring of disease activity. Promising diagnostic autoantibodies include anti-carbamylated proteins and anti-oxidized type II collagen antibodies. Some of the most promising future strategies for redox-based therapeutic compounds are the activation of endogenous cellular antioxidant systems (e.g. Nrf2-dependent pathways), inhibition of disease-relevant sources of ROS/RNS (e.g. isoform-specific NOX inhibitors), or perhaps specifically scavenging disease-related ROS/RNS via site-specific antioxidants.
Collapse
Affiliation(s)
- Miranda J Smallwood
- University of Exeter Medical School, St Luke's Campus, Exeter, Devon EX1 2LU, UK
| | - Ahuva Nissim
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Queen Mary, University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Annie R Knight
- University of Exeter Medical School, St Luke's Campus, Exeter, Devon EX1 2LU, UK
| | - Matthew Whiteman
- University of Exeter Medical School, St Luke's Campus, Exeter, Devon EX1 2LU, UK
| | - Richard Haigh
- University of Exeter Medical School, St Luke's Campus, Exeter, Devon EX1 2LU, UK; Department of Rheumatology, Princess Elizabeth Orthopaedic Centre, Royal Devon and Exeter NHS Foundation Trust (Wonford), Exeter EX2 5DW, UK
| | - Paul G Winyard
- University of Exeter Medical School, St Luke's Campus, Exeter, Devon EX1 2LU, UK.
| |
Collapse
|
19
|
Khan AA, Alsahli MA, Rahmani AH. Myeloperoxidase as an Active Disease Biomarker: Recent Biochemical and Pathological Perspectives. Med Sci (Basel) 2018; 6:medsci6020033. [PMID: 29669993 PMCID: PMC6024665 DOI: 10.3390/medsci6020033] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/04/2018] [Accepted: 04/11/2018] [Indexed: 12/15/2022] Open
Abstract
Myeloperoxidase (MPO) belongs to the family of heme-containing peroxidases, produced mostly from polymorphonuclear neutrophils. The active enzyme (150 kDa) is the product of the MPO gene located on long arm of chromosome 17. The primary gene product undergoes several modifications, such as the removal of introns and signal peptides, and leads to the formation of enzymatically inactive glycosylated apoproMPO which complexes with chaperons, producing inactive proMPO by the insertion of a heme moiety. The active enzyme is a homodimer of heavy and light chain protomers. This enzyme is released into the extracellular fluid after oxidative stress and different inflammatory responses. Myeloperoxidase is the only type of peroxidase that uses H₂O₂ to oxidize several halides and pseudohalides to form different hypohalous acids. So, the antibacterial activities of MPO involve the production of reactive oxygen and reactive nitrogen species. Controlled MPO release at the site of infection is of prime importance for its efficient activities. Any uncontrolled degranulation exaggerates the inflammation and can also lead to tissue damage even in absence of inflammation. Several types of tissue injuries and the pathogenesis of several other major chronic diseases such as rheumatoid arthritis, cardiovascular diseases, liver diseases, diabetes, and cancer have been reported to be linked with MPO-derived oxidants. Thus, the enhanced level of MPO activity is one of the best diagnostic tools of inflammatory and oxidative stress biomarkers among these commonly-occurring diseases.
Collapse
Affiliation(s)
- Amjad A Khan
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, AlQassim, P.O. Box 6699, Buraidah 51452, Saudi Arabia.
| | - Mohammed A Alsahli
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, AlQassim, P.O. Box 6699, Buraidah 51452, Saudi Arabia.
| | - Arshad H Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, AlQassim, P.O. Box 6699, Buraidah 51452, Saudi Arabia.
| |
Collapse
|
20
|
Ray D, Strickland FM, Richardson BC. Oxidative stress and dietary micronutrient deficiencies contribute to overexpression of epigenetically regulated genes by lupus T cells. Clin Immunol 2018; 196:97-102. [PMID: 29654844 DOI: 10.1016/j.clim.2018.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 04/05/2018] [Accepted: 04/05/2018] [Indexed: 11/30/2022]
Abstract
Patients with active lupus have altered T cells characterized by low DNA methyltransferase levels. We hypothesized that low DNA methyltransferase levels synergize with low methionine levels to cause greater overexpression of genes normally suppressed by DNA methylation. CD4+ T cells from lupus patients and controls were stimulated with PHA then cultured in custom media with normal or low methionine levels. Oxidative stress was induced by treating the normal CD4+ T cells with peroxynitrite prior to culture. Methylation sensitive gene expression was measured by flow cytometry. Results showed low methionine levels caused greater overexpression of methylation sensitive genes in peroxynitrite treated T cells relative to untreated T cells, and in T cells from lupus patients relative to T cells from healthy controls. In conclusion, low dietary transmethylation micronutrient levels and low DNA methyltransferase levels caused either by oxidative stress or lupus, have additive effects on methylation sensitive T cell gene expression.
Collapse
Affiliation(s)
- Donna Ray
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Faith M Strickland
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Bruce C Richardson
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
21
|
Protective Effects of Hydroxychloroquine against Accelerated Atherosclerosis in Systemic Lupus Erythematosus. Mediators Inflamm 2018; 2018:3424136. [PMID: 29670462 PMCID: PMC5835241 DOI: 10.1155/2018/3424136] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 12/10/2017] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular (CV) morbidity and mortality are a challenge in management of patients with systemic lupus erythematosus (SLE). Higher risk of CV disease in SLE patients is mostly related to accelerated atherosclerosis. Nevertheless, high prevalence of traditional cardiovascular risk factors in SLE patients does not fully explain the increased CV risk. Despite the pathological bases of accelerated atherosclerosis are not fully understood, it is thought that this process is driven by the complex interplay between SLE and atherosclerosis pathogenesis. Hydroxychloroquine (HCQ) is a cornerstone in treatment of SLE patients and has been thought to exert a broad spectrum of beneficial effects on disease activity, prevention of damage accrual, and mortality. Furthermore, HCQ is thought to protect against accelerated atherosclerosis targeting toll-like receptor signaling, cytokine production, T-cell and monocyte activation, oxidative stress, and endothelial dysfunction. HCQ was also described to have beneficial effects on traditional CV risk factors, such as dyslipidemia and diabetes. In conclusion, despite lacking randomized controlled trials unambiguously proving the protection of HCQ against accelerated atherosclerosis and incidence of CV events in SLE patients, evidence analyzed in this review is in favor of its beneficial effect.
Collapse
|
22
|
Increased lipid and protein oxidation and lowered anti-oxidant defenses in systemic lupus erythematosus are associated with severity of illness, autoimmunity, increased adhesion molecules, and Th1 and Th17 immune shift. Immunol Res 2017; 66:158-171. [DOI: 10.1007/s12026-017-8960-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
23
|
Abstract
Oxidative stress (OS) plays an important role in the pathogenesis of a variety of autoimmune diseases (ADs) and many environmental agents participate in this process. Environmental agents, including trichloroethylene (TCE), silica, pristane, mercury, and smoke, are known to induce an autoimmune response, potentially through OS-mediated mechanisms. Here, we focus on unraveling the targets and signaling pathways that have been mechanistically linked with OS, as a result of exposure to these and numerous other environmental agents, and their impact on the immune system in triggering ADs. Antioxidants and molecular targets impeding autoimmunity by targeting specific signaling pathways are also reviewed. The review not only provides an overview of the current knowledge and evidence showing strong associations between environmental exposures, OS, and ADs, but also plausible mechanisms by which OS causes autoimmunity/ADs. We also discuss areas that require additional approaches, such as unraveling specific events/mechanisms leading to such devastating diseases and measures to prevent or attenuate such diseases.
Collapse
Affiliation(s)
- M Firoze Khan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555
| | - Gangduo Wang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555
| |
Collapse
|
24
|
Effects of oral Lactobacillus administration on antioxidant activities and CD4+CD25+forkhead box P3 (FoxP3)+ T cells in NZB/W F1 mice. Br J Nutr 2017; 118:333-342. [PMID: 28901888 DOI: 10.1017/s0007114517002112] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that is characterised by a dysregulation of the immune system, which causes inflammation responses, excessive oxidative stress and a reduction in the number of cluster of differentiation (CD)4+CD25+forkhead box P3 (FoxP3)+ T cells. Supplementation with certain Lactobacillus strains has been suggested to be beneficial in the comprehensive treatment of SLE. However, little is known about the effect and mechanism of certain Lactobacillus strains on SLE. To investigate the effects of Lactobacillus on SLE, NZB/W F1 mice were orally gavaged with Lactobacillus paracasei GMNL-32 (GMNL-32), Lactobacillus reuteri GMNL-89 (GMNL-89) and L. reuteri GMNL-263 (GMNL-263). Supplementation with GMNL-32, GMNL-89 and GMNL-263 significantly increased antioxidant activity, reduced IL-6 and TNF-α levels and significantly decreased the toll-like receptors/myeloid differentiation primary response gene 88 signalling in NZB/W F1 mice. Notably, supplementation with GMNL-263, but not GMNL-32 and GMNL-89, in NZB/W F1 mice significantly increased the differentiation of CD4+CD25+FoxP3+ T cells. These findings reveal beneficial effects of GMNL-32, GMNL-89 and GMNL-263 on NZB/W F1 mice and suggest that these specific Lactobacillus strains can be used as part of a comprehensive treatment of SLE patients.
Collapse
|
25
|
Al-Griw MA, Treesh SA, Alghazeer RO, Regeai SO. Environmentally toxicant exposures induced intragenerational transmission of liver abnormalities in mice. Open Vet J 2017; 7:244-253. [PMID: 28884077 PMCID: PMC5579565 DOI: 10.4314/ovj.v7i3.8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 07/31/2017] [Indexed: 11/29/2022] Open
Abstract
Environmental toxicants such as chemicals, heavy metals, and pesticides have been shown to promote transgenerational inheritance of abnormal phenotypes and/or diseases to multiple subsequent generations following parental and/or ancestral exposures. This study was designed to examine the potential transgenerational action of the environmental toxicant trichloroethane (TCE) on transmission of liver abnormality, and to elucidate the molecular etiology of hepatocyte cell damage. A total of thirty two healthy immature female albino mice were randomly divided into three equal groups as follows: a sham group, which did not receive any treatment; a vehicle group, which received corn oil alone, and TCE treated group (3 weeks, 100 μg/kg i.p., every 4th day). The F0 and F1 generation control and TCE populations were sacrificed at the age of four months, and various abnormalities histpathologically investigated. Cell death and oxidative stress indices were also measured. The present study provides experimental evidence for the inheritance of environmentally induced liver abnormalities in mice. The results of this study show that exposure to the TCE promoted adult onset liver abnormalities in F0 female mice as well as unexposed F1 generation offspring. It is the first study to report a transgenerational liver abnormalities in the F1 generation mice through maternal line prior to gestation. This finding was based on careful evaluation of liver histopathological abnormalities, apoptosis of hepatocytes, and measurements of oxidative stress biomarkers (lipid peroxidation, protein carbonylation, and nitric oxide) in control and TCE populations. There was an increase in liver histopathological abnormalities, cell death, and oxidative lipid damage in F0 and F1 hepatic tissues of TCE treated group. In conclusion, this study showed that the biological and health impacts of environmental toxicant TCE do not end in maternal adults, but are passed on to offspring generations. Hence, linking observed liver abnormality in the offspring to environmental exposure of their parental line. This study also illustrated that oxidative stress and apoptosis appear to be a molecular component of the hepatocyte cell injury.
Collapse
Affiliation(s)
- Mohamed A Al-Griw
- Developmental Biology Division, Zoology Department, Faculty of Science, University of Tripoli, Tripoli, Libya
| | - Soad A Treesh
- Department of Histology and Medical Genetics, Faculty of Medicine, University of Tripoli, Tripoli, Libya
| | - Rabia O Alghazeer
- Chemistry Department, Faculty of Science, University of Tripoli, Tripoli, Libya
| | - Sassia O Regeai
- Developmental Biology Division, Zoology Department, Faculty of Science, University of Tripoli, Tripoli, Libya
| |
Collapse
|
26
|
Arif Z, Neelofar K, Tarannum A, Arfat MY, Ahmad S, Zaman A, Khan MA, Badar A, Islam SN, Iqubal MA. SLE autoantibodies are well recognized by peroxynitrite-modified-HSA: Its implications in the pathogenesis of SLE. Int J Biol Macromol 2017; 106:1240-1249. [PMID: 28851636 DOI: 10.1016/j.ijbiomac.2017.08.122] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 08/21/2017] [Accepted: 08/22/2017] [Indexed: 01/05/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder where the role of inflammatory processes in the etiopathogenesis is well documented. Despite extensive research, the trigger for initiation of the disease has not been identified. Peroxynitrite, a strong nitrating/oxidizing agent has been reported in SLE and other autoimmune diseases. In this study, human serum albumin (HSA) was exposed to peroxynitrite for 30min at 37°C. The structure of HSA was grossly perturbed when examined by various physico-chemical techniques. Peroxynitrite mediated nitration of HSA was confirmed by LCMS/MS. Furthermore, increase in hydrodynamic radius of peroxynitrite-modified-HSA suggests the attachment of nitro group(s). Aggregation in peroxynitrite-modified-HSA was evident in a TEM scan. Nitration, oxidation, cross linking, aggregation etc conferred immunogenicity on peroxynitrite-modified-HSA. High titre antibodies were elicited in rabbits immunized with peroxynitrite-modified-HSA. Induced antibodies were highly specific for peroxynitrite-modified-HSA but showed considerable binding with other nitrated molecules. Direct binding/inhibition ELISA carried out with autoantibodies in SLE sera showed preferential binding with peroxynitrite-modified-HSA. Anti-nDNA positive IgG from SLE sera showed preference for peroxynitrite-modified-HSA when subjected to immunoassay (direct binding and inhibition) and mobility shift assay. Our results reinforce the role of augmented inflammation in SLE progression.
Collapse
Affiliation(s)
- Zarina Arif
- Dept. of Biochemistry, JN Medical College, Aligarh Muslim University, Aligarh, 202002, India.
| | - Km Neelofar
- Dept. of Biochemistry, JN Medical College, Aligarh Muslim University, Aligarh, 202002, India
| | - Akhlas Tarannum
- Dept. of Biochemistry, JN Medical College, Aligarh Muslim University, Aligarh, 202002, India
| | - Mir Yasir Arfat
- Department of Biotechnology, Islamia College of Science and Commerce, Srinagar (J & K), 190002, India
| | - Shafeeque Ahmad
- Dept. of Biochemistry, JN Medical College, Aligarh Muslim University, Aligarh, 202002, India
| | - Asif Zaman
- Dept. of Biochemistry, JN Medical College, Aligarh Muslim University, Aligarh, 202002, India
| | - Mohd Adnan Khan
- Dept. of Biochemistry, JN Medical College, Aligarh Muslim University, Aligarh, 202002, India
| | - Asim Badar
- Dept. of Biochemistry, JN Medical College, Aligarh Muslim University, Aligarh, 202002, India
| | - Shireen Naaz Islam
- Dept. of Biochemistry, JN Medical College, Aligarh Muslim University, Aligarh, 202002, India
| | - Mohammad Arif Iqubal
- Dept. of Biochemistry, JN Medical College, Aligarh Muslim University, Aligarh, 202002, India
| |
Collapse
|
27
|
Al-Griw MA, Alghazeer RO, Al-Azreg SA, Bennour EM. Cellular and molecular etiology of hepatocyte injury in a murine model of environmentally induced liver abnormality. Open Vet J 2016; 6:150-157. [PMID: 27800299 PMCID: PMC5079423 DOI: 10.4314/ovj.v6i3.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 08/25/2016] [Indexed: 01/10/2023] Open
Abstract
Exposures to a wide variety of environmental substances are negatively associated with many biological cell systems both in humans and rodents. Trichloroethane (TCE), a ubiquitous environmental toxicant, is used in large quantities as a dissolvent, metal degreaser, chemical intermediate, and component of consumer products. This increases the likelihood of human exposure to these compounds through dermal, inhalation and oral routes. The present in vivo study was aimed to investigate the possible cellular and molecular etiology of liver abnormality induced by early exposure to TCE using a murine model. The results showed a significant increase in liver weight. Histopathological examination revealed a TCE-induced hepatotoxicity which appeared as heavily congested central vein and blood sinusoids as well as leukocytic infiltration. Mitotic figures and apoptotic changes such as chromatin condensation and nuclear fragments were also identified. Cell death analysis demonstrates hepatocellular apoptosis was evident in the treated mice compared to control. TCE was also found to induce oxidative stress as indicated by an increase in the levels of lipid peroxidation, an oxidative stress marker. There was also a significant decrease in the DNA content of the hepatocytes of the treated groups compared to control. Agarose gel electrophoresis also provided further biochemical evidence of apoptosis by showing internucleosomal DNA fragmentation in the liver cells, indicating oxidative stress as the cause of DNA damage. These results suggest the need for a complete risk assessment of any new chemical prior to its arrival into the consumer market.
Collapse
Affiliation(s)
- M A Al-Griw
- Division of Developmental Biology, Zoology Department, Faculty of Science, University of Tripoli, Tripoli, Libya
| | - R O Alghazeer
- Chemistry Department, Faculty of Science, University of Tripoli, Tripoli, Libya
| | - S A Al-Azreg
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, University of Tripoli, Tripoli, Libya
| | - E M Bennour
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tripoli, Tripoli, Libya
| |
Collapse
|
28
|
Lee HT, Wu TH, Lin CS, Lee CS, Wei YH, Tsai CY, Chang DM. The pathogenesis of systemic lupus erythematosus - From the viewpoint of oxidative stress and mitochondrial dysfunction. Mitochondrion 2016; 30:1-7. [DOI: 10.1016/j.mito.2016.05.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 05/18/2016] [Accepted: 05/20/2016] [Indexed: 01/26/2023]
|
29
|
Stanisavljevic N, Stojanovich L, Marisavljevic D, Djokovic A, Dopsaj V, Kotur-Stevuljevic J, Martinovic J, Memon L, Radovanovic S, Todic B, Lisulov D. Lipid peroxidation as risk factor for endothelial dysfunction in antiphospholipid syndrome patients. Clin Rheumatol 2016; 35:2485-93. [PMID: 27562033 DOI: 10.1007/s10067-016-3369-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 06/06/2016] [Accepted: 07/25/2016] [Indexed: 01/22/2023]
Abstract
The aim of this study was to evaluate oxidative stress markers and it relations to endothelial damage as risk factor for thrombosis in patients with primary (PAPS) and secondary (SAPS) antiphospholipid syndrome (APS) in correlation to traditional risk factors. Flow-mediated (FMD) and nitroglycerine (NMD)-induced dilation of the brachial artery were studied in 140 APS patients (90 PAPS, 50 SAPS) and 40 controls matched by age, sex, and conventional risk factors for atherosclerosis. Markers of oxidative stress, lipid hydroperoxydes (LOOH), advanced oxidation protein products (AOPP), total sulfhydryl groups (tSHG), and paraoxonase 1 activity (PON1) were determined by spectrophotometric method. Oxidative stress dominates in APS patients. LOOH and AOPP correlate to lipid fractions (p < 0.05), unlike PON1, tSHG that correlated to antiphospholipid antibody positivity (p < 0.05). FMD was lower in APS patients comparing to controls (p < 0.001). Cholesterol is independent variable for FMD impairment in control group (p = 0.011); LOOH in PAPS (p = 0.004); LOOH, aCL, and triglycerides in SAPS patients (p = 0.009, p = 0.049, and p = 0.012, respectively). Combined predictive of aCL and LOOH is better for FMD impairment than LOOH alone in both PAPS and SAPS patients (AUC 0.727, p = 0.001, 95 % CI 0.616-0.837 and AUC 0.824, p˂0.001, 95 % CI 0.690-0.957, respectively). Lipid peroxidation is independent predictor for endothelial dysfunction in APS patients. We demonstrated synergistic effect of aCL and LOOH as risk for endothelial impairment in both PAPS and SAPS patients.
Collapse
Affiliation(s)
- Natasa Stanisavljevic
- University Clinical Center "Bezanijska kosa", Bezanijska kosa bb, Belgrade, 11070, Serbia.
| | - L Stojanovich
- University Clinical Center "Bezanijska kosa", Bezanijska kosa bb, Belgrade, 11070, Serbia
| | - D Marisavljevic
- University Clinical Center "Bezanijska kosa", Bezanijska kosa bb, Belgrade, 11070, Serbia.,Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - A Djokovic
- University Clinical Center "Bezanijska kosa", Bezanijska kosa bb, Belgrade, 11070, Serbia.,Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - V Dopsaj
- Clinical Center of Serbia, Faculty of Pharmacy Belgrade, University of Belgrade, Belgrade, Serbia
| | - J Kotur-Stevuljevic
- Institute of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - J Martinovic
- Rakovica Community Health Center, Belgrade, Serbia
| | - L Memon
- University Clinical Center "Bezanijska kosa", Bezanijska kosa bb, Belgrade, 11070, Serbia
| | - S Radovanovic
- University Clinical Center "Bezanijska kosa", Bezanijska kosa bb, Belgrade, 11070, Serbia
| | - B Todic
- University Clinical Center "Bezanijska kosa", Bezanijska kosa bb, Belgrade, 11070, Serbia
| | - D Lisulov
- University Clinical Center "Bezanijska kosa", Bezanijska kosa bb, Belgrade, 11070, Serbia
| |
Collapse
|
30
|
Dal S, Sigrist S. The Protective Effect of Antioxidants Consumption on Diabetes and Vascular Complications. Diseases 2016; 4:E24. [PMID: 28933404 PMCID: PMC5456287 DOI: 10.3390/diseases4030024] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/20/2016] [Accepted: 06/23/2016] [Indexed: 12/14/2022] Open
Abstract
Obesity and diabetes is generally accompanied by a chronic state of oxidative stress, disequilibrium in the redox balance, implicated in the development and progression of complications such as micro- and macro-angiopathies. Disorders in the inner layer of blood vessels, the endothelium, play an early and critical role in the development of these complications. Blunted endothelium-dependent relaxation and/or contractions are quietly associated to oxidative stress. Thus, preserving endothelial function and oxidative stress seems to be an optimization strategy in the prevention of vascular complications associated with diabetes. Diet is a major lifestyle factor that can greatly influence the incidence and the progression of type 2 diabetes and cardiovascular complications. The notion that foods not only provide basic nutrition but can also prevent diseases and ensure good health and longevity is now attained greater prominence. Some dietary and lifestyle modifications associated to antioxidative supply could be an effective prophylactic means to fight against oxidative stress in diabesity and complications. A significant benefit of phytochemicals (polyphenols in wine, grape, teas), vitamins (ascorbate, tocopherol), minerals (selenium, magnesium), and fruits and vegetables in foods is thought to be capable of scavenging free radicals, lowering the incidence of chronic diseases. In this review, we discuss the role of oxidative stress in diabetes and complications, highlight the endothelial dysfunction, and examine the impact of antioxidant foods, plants, fruits, and vegetables, currently used medication with antioxidant properties, in relation to the development and progression of diabetes and cardiovascular complications.
Collapse
Affiliation(s)
- Stéphanie Dal
- DIATHEC EA 7294 UMR Centre Européen d'Etude du Diabète (CeeD), Université de Strasbourg (UdS), boulevard René Leriche, Strasbourg 67200, France.
| | - Séverine Sigrist
- DIATHEC EA 7294 UMR Centre Européen d'Etude du Diabète (CeeD), Université de Strasbourg (UdS), boulevard René Leriche, Strasbourg 67200, France.
| |
Collapse
|
31
|
Lopez LR, Salazar-Paramo M, Palafox-Sanchez C, Hurley BL, Matsuura E, Garcia-De La Torre I. Oxidized low-density lipoprotein and β2-glycoprotein I in patients with systemic lupus erythematosus and increased carotid intima-media thickness: implications in autoimmune-mediated atherosclerosis. Lupus 2016; 15:80-6. [PMID: 16539278 DOI: 10.1191/0961203306lu2267oa] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Oxidative stress and LDL modification (oxLDL) are early pro-atherogenic events. OxLDL binds β2GPI producing immunogenic oxLDL/β2GPI complexes. Antibodies to these complexes have been associated with arterial thrombosis in patients with systemic lupus erythematosus (SLE) and antiphospholipid syndrome (APS). Circulating oxLDL/β2GPI complexes, IgG and IgM antibodies to these complexes were measured by ELISA in 30 SLE patients asymptomatic for cardiovascular disease (mean age 31 years) and 27 age/sex matched healthy controls. Carotid intima-media thickness (IMT) was measured by ultrasound in all patients and controls. Forty-seven percent of SLE presented plaques (median IMT of 0.65 ± 0.12 mm) while only 7% of the controls had plaques (median IMT of 0.50 ± 0.04 mm, P < 0.001). Median optical density (OD450nm) for oxLDL/β2GPI complexes in SLE was 0.244 ± 0.07, higher than controls (0.174 ± 0.09, P < 0.001). Median OD for IgG anti-oxLDL/β2GPI antibodies was also higher in SLE (0.297 ± 0.26) compared to controls (0.194 ± 0.07, P < 0.001) while the median OD for IgM antibodies in SLE (0.444 ± 0.46) was not different than controls (0.326 ± 0.22, P = 0.267). There was no correlation between IMT and oxLDL/β2GPI complexes, IgG or IgM antibodies, possibly reflecting the complex interrelationship between these serologic elements and tissue factors in the arterial wall. These results support the hypothesis that oxLDL/β2GPI complexes and IgG (not IgM) anti-oxLDL/β2GPI antibodies contribute to the development of autoimmune-mediated atherosclerosis
Collapse
Affiliation(s)
- L R Lopez
- Corgenix, Inc., Westminster, Colorado, USA.
| | | | | | | | | | | |
Collapse
|
32
|
Kienhöfer D, Boeltz S, Hoffmann MH. Reactive oxygen homeostasis – the balance for preventing autoimmunity. Lupus 2016; 25:943-54. [DOI: 10.1177/0961203316640919] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Being mainly known for their role in the antimicrobial defense and collateral damage they cause in tissues as agents of oxidative stress, reactive oxygen species were considered “the bad guys” for decades. However, in the last years it was shown that the absence of reactive oxygen species can lead to the development of immune-mediated inflammatory diseases. Animal models of lupus, arthritis and psoriasis revealed reactive oxygen species-deficiency as a potent driver of pathogenesis. On the contrary, in chronic stages oxidative stress can still contribute to progression of inflammation. It seems that a neatly adjusted redox balance is necessary to sustain an immune state that both prevents the development of overt autoimmunity and attenuates chronic stages of disease.
Collapse
Affiliation(s)
- D Kienhöfer
- Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Department of Internal Medicine 3—Rheumatology and Immunology, Erlangen, Germany
| | - S Boeltz
- Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Department of Internal Medicine 3—Rheumatology and Immunology, Erlangen, Germany
| | - M H Hoffmann
- Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Department of Internal Medicine 3—Rheumatology and Immunology, Erlangen, Germany
| |
Collapse
|
33
|
Prooxidant-Antioxidant Balance in Patients with Systemic Lupus Erythematosus and Its Relationship with Clinical and Laboratory Findings. Autoimmune Dis 2016; 2016:4343514. [PMID: 26925260 PMCID: PMC4746302 DOI: 10.1155/2016/4343514] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 01/11/2016] [Indexed: 01/29/2023] Open
Abstract
Aim. This study was aimed at evaluating prooxidant-antioxidant balance (PAB) in patients with systemic lupus erythematosus (SLE) and its relationship with laboratory findings and clinical manifestations. Methods. In this case-control study, 60 patients with SLE and 60 healthy individuals were enrolled. The blood samples were collected and their sera were separated. Subsequently, the prooxidant-antioxidant balance value was evaluated using PAB assay for each sample. Results. The mean of PAB values in SLE patients was significantly higher than healthy controls (147.3 ± 42 versus 84.8 ± 32.2 HK, P < 0.0001). Furthermore, in SLE patients, there was a positive significant correlation between the PAB and erythrocyte sedimentation rate (ESR) (r = 0.492, P < 0.001). In addition, the PAB values in patients with alopecia, discoid rash, oral ulcers, arthritis, and nephritis were significantly higher than those without these manifestations. Conclusion. The findings of current study showed that the mean of PAB was significantly higher in SLE patients and PAB was correlated with ESR. Moreover increased PAB was found in SLE patients with alopecia, discoid rash, oral ulcers, arthritis, and nephritis. These findings suggest that the measurement of PAB may be useful to show oxidative stress condition in SLE patients.
Collapse
|
34
|
Bai MJ, Feng J, Yu F, Yan CL, Cui CJ, Huang L, Feng ZR. Serum Myeloperoxidase Level in Systemic Lupus Erythematosus. ACTA ACUST UNITED AC 2016; 30:199-202. [PMID: 26564422 DOI: 10.1016/s1001-9294(15)30048-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
35
|
Robbins KS, Gong Y, Wells ML, Greenspan P, Pegg RB. Reprint of “Investigation of the antioxidant capacity and phenolic constituents of U.S. pecans”. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.05.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
36
|
Kurien BT, Harris VM, Quadri SMS, Coutinho-de Souza P, Cavett J, Moyer A, Ittiq B, Metcalf A, Ramji HF, Truong D, Kumar R, Koelsch KA, Centola M, Payne A, Danda D, Scofield RH. Significantly reduced lymphadenopathy, salivary gland infiltrates and proteinuria in MRL-lpr/lpr mice treated with ultrasoluble curcumin/turmeric: increased survival with curcumin treatment. Lupus Sci Med 2015; 2:e000114. [PMID: 26380101 PMCID: PMC4567741 DOI: 10.1136/lupus-2015-000114] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 08/05/2015] [Accepted: 08/20/2015] [Indexed: 11/26/2022]
Abstract
Objectives Commercial curcumin (CU), derived from food spice turmeric (TU), has been widely studied as a potential therapeutic for a variety of oncological and inflammatory conditions. Lack of solubility/bioavailability has hindered curcumin's therapeutic efficacy in human diseases. We have solubilised curcumin in water applying heat/pressure, obtaining up to 35-fold increase in solubility (ultrasoluble curcumin (UsC)). We hypothesised that UsC or ultrasoluble turmeric (UsT) will ameliorate systemic lupus erythematosus (SLE) and Sjögren's syndrome (SS)-like disease in MRL-lpr/lpr mice. Methods Eighteen female MRL-lpr/lpr (6 weeks old) and 18 female MRL-MpJ mice (6 weeks old) were used. Female MRL-lpr/lpr mice develop lupus-like disease at the 10th week and die at an average age of 17 weeks. MRL-MpJ mice develop lupus-like disease around 47 weeks and typically die at 73 weeks. Six mice of each strain received autoclaved water only (lpr-water or MpJ-water group), UsC (lpr-CU or MpJ-CU group) or UsT (lpr-TU or MpJ-TU group) in the water bottle. Results UsC or UsT ameliorates SLE in the MRL-lpr/lpr mice by significantly reducing lymphoproliferation, proteinuria, lesions (tail) and autoantibodies. lpr-CU group had a 20% survival advantage over lpr-water group. However, lpr-TU group lived an average of 16 days shorter than lpr-water group due to complications unrelated to lupus-like illness. CU/TU treatment inhibited lymphadenopathy significantly compared with lpr-water group (p=0.03 and p=0.02, respectively) by induction of apoptosis. Average lymph node weights were 2606±1147, 742±331 and 385±68 mg, respectively, for lpr-water, lpr-CU and lpr-TU mice. Transferase dUTP nick end labelling assay showed that lymphocytes in lymph nodes of lpr-CU and lpr-TU mice underwent apoptosis. Significantly reduced cellular infiltration of the salivary glands in the lpr-TU group compared with the lpr-water group, and a trend towards reduced kidney damage was observed in the lpr-CU and lpr-TU groups. Conclusions These studies show that UsC/UsT could prove useful as a therapeutic intervention in SLE/SS.
Collapse
Affiliation(s)
- Biji T Kurien
- Department of Medicine , University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma , USA ; Oklahoma Medical Research Foundation , Arthritis & Clinical Immunology Program , Oklahoma City, Oklahoma , USA ; Department Veterans Affairs Medical Center, Oklahoma City, Oklahoma, USA
| | - Valerie M Harris
- Department of Medicine , University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma , USA ; Oklahoma Medical Research Foundation , Arthritis & Clinical Immunology Program , Oklahoma City, Oklahoma , USA
| | - Syed M S Quadri
- Department of Medicine , University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma , USA ; Oklahoma Medical Research Foundation , Arthritis & Clinical Immunology Program , Oklahoma City, Oklahoma , USA
| | - Patricia Coutinho-de Souza
- Oklahoma Medical Research Foundation , Arthritis & Clinical Immunology Program , Oklahoma City, Oklahoma , USA
| | - Joshua Cavett
- Department of Medicine , University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma , USA ; Oklahoma Medical Research Foundation , Arthritis & Clinical Immunology Program , Oklahoma City, Oklahoma , USA
| | - Amanda Moyer
- Department of Medicine , University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma , USA
| | - Bilal Ittiq
- Department of Medicine , University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma , USA
| | - Angela Metcalf
- Oklahoma Medical Research Foundation , Arthritis & Clinical Immunology Program , Oklahoma City, Oklahoma , USA
| | - Husayn F Ramji
- Oklahoma School of Science and Mathematics, Oklahoma City, Oklahoma, USA ; University of Oklahoma , Norman, Oklahoma , USA
| | - Dat Truong
- Oklahoma School of Science and Mathematics, Oklahoma City, Oklahoma, USA
| | - Ramesh Kumar
- Oklahoma Medical Research Foundation , Arthritis & Clinical Immunology Program , Oklahoma City, Oklahoma , USA
| | - Kristi A Koelsch
- Department of Medicine , University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma , USA ; Oklahoma Medical Research Foundation , Arthritis & Clinical Immunology Program , Oklahoma City, Oklahoma , USA ; Department Veterans Affairs Medical Center, Oklahoma City, Oklahoma, USA
| | - Mike Centola
- Haus Bioceuticals , Oklahoma City, Oklahoma , USA
| | - Adam Payne
- Haus Bioceuticals , Oklahoma City, Oklahoma , USA
| | | | - R Hal Scofield
- Department of Medicine , University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma , USA ; Oklahoma Medical Research Foundation , Arthritis & Clinical Immunology Program , Oklahoma City, Oklahoma , USA ; Department Veterans Affairs Medical Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
37
|
Achilli C, Ciana A, Minetti G. The discovery of methionine sulfoxide reductase enzymes: An historical account and future perspectives. Biofactors 2015; 41:135-52. [PMID: 25963551 DOI: 10.1002/biof.1214] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 04/19/2015] [Indexed: 01/26/2023]
Abstract
L-Methionine (L-Met) is the only sulphur-containing proteinogenic amino acid together with cysteine. Its importance is highlighted by it being the initiator amino acid for protein synthesis in all known living organisms. L-Met, free or inserted into proteins, is sensitive to oxidation of its sulfide moiety, with formation of L-Met sulfoxide. The sulfoxide could not be inserted into proteins, and the oxidation of L-Met in proteins often leads to the loss of biological activity of the affected molecule. Key discoveries revealed the existence, in rats, of a metabolic pathway for the reduction of free L-Met sulfoxide and, later, in Escherichia coli, of the enzymatic reduction of L-Met sulfoxide inserted in proteins. Upon oxidation, the sulphur atom becomes a new stereogenic center, and two stable diastereoisomers of L-Met sulfoxide exist. A fundamental discovery revealed the existence of two unrelated families of enzymes, MsrA and MsrB, whose members display opposite stereospecificity of reduction for the two sulfoxides. The importance of Msrs is additionally emphasized by the discovery that one of the only 25 selenoproteins expressed in humans is a Msr. The milestones on the road that led to the discovery and characterization of this group of antioxidant enzymes are recounted in this review.
Collapse
Affiliation(s)
- Cesare Achilli
- Laboratories of Biochemistry, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Annarita Ciana
- Laboratories of Biochemistry, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Giampaolo Minetti
- Laboratories of Biochemistry, Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| |
Collapse
|
38
|
|
39
|
Houée-Lévin C, Bobrowski K, Horakova L, Karademir B, Schöneich C, Davies MJ, Spickett CM. Exploring oxidative modifications of tyrosine: An update on mechanisms of formation, advances in analysis and biological consequences. Free Radic Res 2015; 49:347-73. [DOI: 10.3109/10715762.2015.1007968] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
40
|
Dey-Rao R, Sinha AA. Genome-wide transcriptional profiling of chronic cutaneous lupus erythematosus (CCLE) peripheral blood identifies systemic alterations relevant to the skin manifestation. Genomics 2014; 105:90-100. [PMID: 25451738 DOI: 10.1016/j.ygeno.2014.11.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 10/24/2014] [Accepted: 11/11/2014] [Indexed: 12/15/2022]
Abstract
Major gaps remain regarding pathogenetic mechanisms underlying clinical heterogeneity in lupus erythematosus (LE). As systemic changes are likely to underlie skin specific manifestation, we analyzed global gene expression in peripheral blood of a small cohort of chronic cutaneous LE (CCLE) patients and healthy individuals. Unbiased hierarchical clustering distinguished patients from controls revealing a "disease" based signature. Functional annotation of the differentially expressed genes (DEGs) highlight enrichment of interferon related immune response and apoptosis signatures, along with other key pathways. There is a 26% overlap of the blood and lesional skin transcriptional profile from a previous analysis by our group. We identified four transcriptional "hot spots" at chromosomal regions harboring statistically increased numbers of DEGs which offer prioritized potential loci for downstream fine mapping studies in the search for CCLE specific susceptibility loci. Additionally, we uncover evidence to support both shared and distinct mechanisms for cutaneous and systemic manifestations of lupus.
Collapse
Affiliation(s)
- R Dey-Rao
- Department of Dermatology, University at Buffalo, Buffalo, NY, USA
| | - A A Sinha
- Department of Dermatology, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
41
|
Wang W, Jian Z, Guo J, Ning X. Increased levels of serum myeloperoxidase in patients with active rheumatoid arthritis. Life Sci 2014; 117:19-23. [DOI: 10.1016/j.lfs.2014.09.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 09/12/2014] [Accepted: 09/13/2014] [Indexed: 12/15/2022]
|
42
|
McPherson PAC, Türemen BT. 3,4-Dihydroxy-l-phenylalanine as a biomarker of oxidative damage in proteins: improved detection using cloud-point extraction and HPLC. Biochem Biophys Res Commun 2014; 452:376-81. [PMID: 25157806 DOI: 10.1016/j.bbrc.2014.08.080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Accepted: 08/16/2014] [Indexed: 10/24/2022]
Abstract
Oxidized protein adducts are formed under conditions of oxidative stress and may represent a valuable biomarker for a variety of diseases which share this common aetiology. A suitable candidate biomarker for oxidized proteins is protein-bound 3,4-dihydroxyl-l-phenylalanine (l-DOPA), which is formed on 3'-hydroxylation of tyrosine residues by hydroxyl radicals. Existing methodologies to measure protein-bound l-DOPA employ lengthy acid hydrolysis steps (ca. 16h) which may cause artifactual protein oxidation, followed by HPLC with detection based on the intrinsic fluorescence of l-DOPA. We report a novel method for the measurement of protein-bound l-DOPA which involves rapid hydrolysis followed by pre-column concentration of 6-aminoquinolyl-derivatives using cloud-point extraction. The derivatized material is resolved by reversed-phase HPLC in less than 30min and has derivatization chemistry compatible with both UV and fluorescent detection, providing detection down to the femtomole level. The method provides identical results to those found with highly specific ELISA-based techniques and requires only basic instrumentation. The stability of the 6-aminoquinolyl-derivatives together with the fast and sensitive nature of the assay will be appealing to those who require large sample throughput.
Collapse
Affiliation(s)
| | - Bryn T Türemen
- School of Applied Science, Belfast Metropolitan College, Belfast, UK
| |
Collapse
|
43
|
Detection and characterization of autoantibodies against modified self-proteins in SLE sera after exposure to reactive oxygen and nitrogen species. Methods Mol Biol 2014; 1134:163-71. [PMID: 24497361 DOI: 10.1007/978-1-4939-0326-9_12] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
There are over 120 types of autoantibodies found in the blood of SLE patients against cellular and extracellular components in both their native and posttranslationally modified forms. In recent years, these autoantibodies have provoked interest as initiators of pathology and as biomarkers of disease activity. Often, the host antigens employed in lab-based and commercially developed immunoassays use non-human antigen or non-modified host antigen as a probe for autoantibodies. Here, we describe methods to posttranslationally modify host antigens, which better represent the antigen recognized by autoantibodies in vivo. This has implications in developing immunoassay with greater sensitivity and specificity.
Collapse
|
44
|
Li Y, Gorelik G, Strickland FM, Richardson BC. Oxidative stress, T cell DNA methylation, and lupus. Arthritis Rheumatol 2014; 66:1574-82. [PMID: 24577881 PMCID: PMC4141415 DOI: 10.1002/art.38427] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 02/13/2014] [Indexed: 12/30/2022]
Abstract
Objective Lupus develops when genetically predisposed people encounter environmental agents, such as ultraviolet light, silica, infections, and cigarette smoke, that cause oxidative stress, but how oxidative damage modifies the immune system to cause lupus flares is unknown. We previously showed that inhibiting DNA methylation in CD4+ T cells by blocking ERK pathway signaling is sufficient to alter gene expression, and that the modified cells cause lupus-like autoimmunity in mice. We also reported that T cells from patients with active lupus have decreased ERK pathway signaling, have decreased DNA methylation, and overexpress genes normally suppressed by DNA methylation. This study was undertaken to test whether oxidizing agents decrease ERK pathway signaling in T cells, decrease DNA methyltransferase levels, and cause demethylation and overexpression of T cell genes similar to that found in T cells from patients with active lupus. Methods CD4+ T cells were treated with the oxidizers H2O2 or ONOO−. Effects on ERK pathway signaling were measured by immunoblotting, DNA methyltransferase 1 (DNMT-1) levels were measured by reverse transcriptase–polymerase chain reaction (RT-PCR), and the methylation and expression of T cell genes were measured using flow cytometry, RT-PCR, and bisulfite sequencing. Results H2O2 and ONOO− inhibited ERK pathway signaling in T cells by inhibiting the upstream regulator protein kinase Cδ, decreased DNMT-1 levels, and caused demethylation and overexpression of genes previously shown to be suppressed by DNA methylation in T cells from patients with active lupus. Conclusion Our findings indicate that oxidative stress may contribute to human lupus flares by inhibiting ERK pathway signaling in T cells to decrease DNMT-1 and cause DNA demethylation.
Collapse
|
45
|
Mahieu MA, Guild CP, Albert CJ, Kondos GT, Carr JJ, Edmundowicz D, Ford DA, Ramsey-Goldman R. Alpha-chlorofatty Acid and coronary artery or aorta calcium scores in women with systemic lupus erythematosus. A pilot study. J Rheumatol 2014; 41:1834-42. [PMID: 25086078 DOI: 10.3899/jrheum.131361] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Alpha-chlorofatty acid (α-ClFA) is one product of myeloperoxidase activity in vivo during atherogenesis and may be a biomarker for cardiovascular disease (CVD). We investigated if serum α-ClFA is associated with subclinical CVD as measured by coronary artery and aorta calcium scores (CAC and AC, respectively) in women with and without systemic lupus erythematosus (SLE). METHODS This pilot project analyzed baseline data from 173 women with SLE and 186 women without SLE participating in a 5-year longitudinal investigation of the Study of Lupus Vascular and Bone Long-term Endpoints (SOLVABLE). Data collection included demographic information, CVD and SLE risk factors, and laboratory assessments. Alpha-ClFA was measured in stored serum by liquid chromatography-mass spectrometry. CAC and AC were measured by computed tomography. Outcome measures were CAC and AC present (CAC > 0 or AC > 0) versus absent (CAC = 0 or AC = 0). Associations between risk factors and CAC or AC were tested with descriptive statistics and multivariate analyses. RESULTS Women with SLE had higher α-ClFA levels than women without SLE (42.0 fmol/25 µl ± 37.3 vs 34.5 fmol/25 µl ± 21.9; p = 0.020). In analyses including individual CVD risk factors, having SLE was independently associated with the presence of CAC (OR 3.42, 95% CI 1.72 to 6.78) but not AC. Alpha-ClFA was not associated with the presence of CAC or AC in patients with SLE. CONCLUSION SLE, but not serum α-ClFA, was associated with the presence of CAC in this pilot project.
Collapse
Affiliation(s)
- Mary A Mahieu
- From the Department of Medicine, Division of Rheumatology, and Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Departments of Pediatrics and Center for Outcomes Research and of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University, Saint Louis, Missouri; Department of Medicine, Section of Cardiology, University of Illinois at Chicago College of Medicine, Chicago, Illinois; and Department of Medicine, Section of Cardiology, Temple University School of Medicine, Philadelphia, Pennsylvania, USA.M.A. Mahieu, MD, Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine; C.P. Guild, MPH, Department of Pediatrics and Center for Outcomes Research, Saint Louis University School of Medicine; C.J. Albert, BA, Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University School of Medicine; G.T. Kondos, MD, Department of Medicine, Section of Cardiology, University of Illinois Chicago College of Medicine; J.J. Carr, MD, Department of Radiology, Northwestern University Feinberg School of Medicine; D. Edmundowicz, MD, Department of Medicine, Section of Cardiology, Temple University School of Medicine; D.A. Ford, PhD, Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University School of Medicine; R. Ramsey-Goldman, MD, DrPH, Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine.
| | - Camelia P Guild
- From the Department of Medicine, Division of Rheumatology, and Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Departments of Pediatrics and Center for Outcomes Research and of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University, Saint Louis, Missouri; Department of Medicine, Section of Cardiology, University of Illinois at Chicago College of Medicine, Chicago, Illinois; and Department of Medicine, Section of Cardiology, Temple University School of Medicine, Philadelphia, Pennsylvania, USA.M.A. Mahieu, MD, Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine; C.P. Guild, MPH, Department of Pediatrics and Center for Outcomes Research, Saint Louis University School of Medicine; C.J. Albert, BA, Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University School of Medicine; G.T. Kondos, MD, Department of Medicine, Section of Cardiology, University of Illinois Chicago College of Medicine; J.J. Carr, MD, Department of Radiology, Northwestern University Feinberg School of Medicine; D. Edmundowicz, MD, Department of Medicine, Section of Cardiology, Temple University School of Medicine; D.A. Ford, PhD, Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University School of Medicine; R. Ramsey-Goldman, MD, DrPH, Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine
| | - Carolyn J Albert
- From the Department of Medicine, Division of Rheumatology, and Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Departments of Pediatrics and Center for Outcomes Research and of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University, Saint Louis, Missouri; Department of Medicine, Section of Cardiology, University of Illinois at Chicago College of Medicine, Chicago, Illinois; and Department of Medicine, Section of Cardiology, Temple University School of Medicine, Philadelphia, Pennsylvania, USA.M.A. Mahieu, MD, Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine; C.P. Guild, MPH, Department of Pediatrics and Center for Outcomes Research, Saint Louis University School of Medicine; C.J. Albert, BA, Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University School of Medicine; G.T. Kondos, MD, Department of Medicine, Section of Cardiology, University of Illinois Chicago College of Medicine; J.J. Carr, MD, Department of Radiology, Northwestern University Feinberg School of Medicine; D. Edmundowicz, MD, Department of Medicine, Section of Cardiology, Temple University School of Medicine; D.A. Ford, PhD, Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University School of Medicine; R. Ramsey-Goldman, MD, DrPH, Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine
| | - George T Kondos
- From the Department of Medicine, Division of Rheumatology, and Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Departments of Pediatrics and Center for Outcomes Research and of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University, Saint Louis, Missouri; Department of Medicine, Section of Cardiology, University of Illinois at Chicago College of Medicine, Chicago, Illinois; and Department of Medicine, Section of Cardiology, Temple University School of Medicine, Philadelphia, Pennsylvania, USA.M.A. Mahieu, MD, Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine; C.P. Guild, MPH, Department of Pediatrics and Center for Outcomes Research, Saint Louis University School of Medicine; C.J. Albert, BA, Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University School of Medicine; G.T. Kondos, MD, Department of Medicine, Section of Cardiology, University of Illinois Chicago College of Medicine; J.J. Carr, MD, Department of Radiology, Northwestern University Feinberg School of Medicine; D. Edmundowicz, MD, Department of Medicine, Section of Cardiology, Temple University School of Medicine; D.A. Ford, PhD, Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University School of Medicine; R. Ramsey-Goldman, MD, DrPH, Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine
| | - James J Carr
- From the Department of Medicine, Division of Rheumatology, and Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Departments of Pediatrics and Center for Outcomes Research and of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University, Saint Louis, Missouri; Department of Medicine, Section of Cardiology, University of Illinois at Chicago College of Medicine, Chicago, Illinois; and Department of Medicine, Section of Cardiology, Temple University School of Medicine, Philadelphia, Pennsylvania, USA.M.A. Mahieu, MD, Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine; C.P. Guild, MPH, Department of Pediatrics and Center for Outcomes Research, Saint Louis University School of Medicine; C.J. Albert, BA, Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University School of Medicine; G.T. Kondos, MD, Department of Medicine, Section of Cardiology, University of Illinois Chicago College of Medicine; J.J. Carr, MD, Department of Radiology, Northwestern University Feinberg School of Medicine; D. Edmundowicz, MD, Department of Medicine, Section of Cardiology, Temple University School of Medicine; D.A. Ford, PhD, Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University School of Medicine; R. Ramsey-Goldman, MD, DrPH, Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine
| | - Daniel Edmundowicz
- From the Department of Medicine, Division of Rheumatology, and Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Departments of Pediatrics and Center for Outcomes Research and of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University, Saint Louis, Missouri; Department of Medicine, Section of Cardiology, University of Illinois at Chicago College of Medicine, Chicago, Illinois; and Department of Medicine, Section of Cardiology, Temple University School of Medicine, Philadelphia, Pennsylvania, USA.M.A. Mahieu, MD, Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine; C.P. Guild, MPH, Department of Pediatrics and Center for Outcomes Research, Saint Louis University School of Medicine; C.J. Albert, BA, Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University School of Medicine; G.T. Kondos, MD, Department of Medicine, Section of Cardiology, University of Illinois Chicago College of Medicine; J.J. Carr, MD, Department of Radiology, Northwestern University Feinberg School of Medicine; D. Edmundowicz, MD, Department of Medicine, Section of Cardiology, Temple University School of Medicine; D.A. Ford, PhD, Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University School of Medicine; R. Ramsey-Goldman, MD, DrPH, Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine
| | - David A Ford
- From the Department of Medicine, Division of Rheumatology, and Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Departments of Pediatrics and Center for Outcomes Research and of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University, Saint Louis, Missouri; Department of Medicine, Section of Cardiology, University of Illinois at Chicago College of Medicine, Chicago, Illinois; and Department of Medicine, Section of Cardiology, Temple University School of Medicine, Philadelphia, Pennsylvania, USA.M.A. Mahieu, MD, Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine; C.P. Guild, MPH, Department of Pediatrics and Center for Outcomes Research, Saint Louis University School of Medicine; C.J. Albert, BA, Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University School of Medicine; G.T. Kondos, MD, Department of Medicine, Section of Cardiology, University of Illinois Chicago College of Medicine; J.J. Carr, MD, Department of Radiology, Northwestern University Feinberg School of Medicine; D. Edmundowicz, MD, Department of Medicine, Section of Cardiology, Temple University School of Medicine; D.A. Ford, PhD, Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University School of Medicine; R. Ramsey-Goldman, MD, DrPH, Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine
| | - Rosalind Ramsey-Goldman
- From the Department of Medicine, Division of Rheumatology, and Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Departments of Pediatrics and Center for Outcomes Research and of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University, Saint Louis, Missouri; Department of Medicine, Section of Cardiology, University of Illinois at Chicago College of Medicine, Chicago, Illinois; and Department of Medicine, Section of Cardiology, Temple University School of Medicine, Philadelphia, Pennsylvania, USA.M.A. Mahieu, MD, Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine; C.P. Guild, MPH, Department of Pediatrics and Center for Outcomes Research, Saint Louis University School of Medicine; C.J. Albert, BA, Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University School of Medicine; G.T. Kondos, MD, Department of Medicine, Section of Cardiology, University of Illinois Chicago College of Medicine; J.J. Carr, MD, Department of Radiology, Northwestern University Feinberg School of Medicine; D. Edmundowicz, MD, Department of Medicine, Section of Cardiology, Temple University School of Medicine; D.A. Ford, PhD, Department of Biochemistry and Molecular Biology, and Center for Cardiovascular Research, Saint Louis University School of Medicine; R. Ramsey-Goldman, MD, DrPH, Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine
| |
Collapse
|
46
|
López-López L, Nieves-Plaza M, Castro MDR, Font YM, Torres-Ramos CA, Vilá LM, Ayala-Peña S. Mitochondrial DNA damage is associated with damage accrual and disease duration in patients with systemic lupus erythematosus. Lupus 2014; 23:1133-41. [PMID: 24899636 DOI: 10.1177/0961203314537697] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE To determine the extent of mitochondrial DNA (mtDNA) damage in systemic lupus erythematosus (SLE) patients compared to healthy subjects and to determine the factors associated with mtDNA damage among SLE patients. METHODS A cross-sectional study was performed in 86 SLE patients (per American College of Rheumatology classification criteria) and 86 healthy individuals matched for age and gender. Peripheral blood mononuclear cells (PBMCs) were collected from subjects to assess the relative amounts of mtDNA damage. Quantitative polymerase chain reaction assay was used to measure the frequency of mtDNA lesions and mtDNA abundance. Socioeconomic-demographic features, clinical manifestations, pharmacologic treatment, disease activity, and damage accrual were determined. Statistical analyses were performed using t test, pairwise correlation, and Pearson's chi-square test (or Fisher's exact test) as appropriate. RESULTS Among SLE patients, 93.0% were women. The mean (SD) age was 38.0 (10.4) years and the mean (SD) disease duration was 8.7 (7.5) years. SLE patients exhibited increased levels of mtDNA damage as shown by higher levels of mtDNA lesions and decreased mtDNA abundance as compared to healthy individuals. There was a negative correlation between disease damage and mtDNA abundance and a positive correlation between mtDNA lesions and disease duration. No association was found between disease activity and mtDNA damage. CONCLUSION PBMCs from SLE patients exhibited more mtDNA damage compared to healthy subjects. Higher levels of mtDNA damage were observed among SLE patients with major organ involvement and damage accrual. These results suggest that mtDNA damage have a potential role in the pathogenesis of SLE.
Collapse
Affiliation(s)
- L López-López
- Department of Medicine (Division of Rheumatology), University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - M Nieves-Plaza
- Puerto Rico Clinical and Translational Research Consortium, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico School of Medicine, Institute for Clinical Research Education, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - M del R Castro
- Department of Pharmacology and Toxicology, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - Y M Font
- Department of Medicine (Division of Rheumatology), University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - C A Torres-Ramos
- Department of Physiology and Biophysics, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - L M Vilá
- Department of Medicine (Division of Rheumatology), University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - S Ayala-Peña
- Department of Pharmacology and Toxicology, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| |
Collapse
|
47
|
Ryan BJ, Nissim A, Winyard PG. Oxidative post-translational modifications and their involvement in the pathogenesis of autoimmune diseases. Redox Biol 2014; 2:715-24. [PMID: 24955328 PMCID: PMC4062766 DOI: 10.1016/j.redox.2014.05.004] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Revised: 05/22/2014] [Accepted: 05/23/2014] [Indexed: 02/07/2023] Open
Abstract
Tissue inflammation results in the production of numerous reactive oxygen, nitrogen and chlorine species, in addition to the products of lipid and sugar oxidation. Some of these products are capable of chemically modifying amino acids. This in turn results in changes to the structure and function of proteins. Increasing evidence demonstrates that such oxidative post-translational modifications result in the generation of neo-epitopes capable of eliciting both innate and adaptive immune responses. In this paper, we focus on how free radicals and related chemical species generated in inflammatory environments modulate the antigenicity of self-proteins, resulting in immune responses which involve the generation of autoantibodies against key autoantigens in autoimmune diseases. As examples, we will focus on Ro-60 and C1q in systemic lupus erythematosus, along with type-II collagen in rheumatoid arthritis. This review also covers some of the emerging literature which demonstrates that neo-epitopes generated by oxidation are conserved, as exemplified by the evolutionarily conserved pathogen-associated molecular patterns (PAMPs). We discuss how these observations relate to the pathogenesis of both human autoimmune diseases and inflammatory disease, such as atherosclerosis. The potential for these neo-epitopes and the immune responses against them to act as biomarkers or therapeutic targets is also discussed. Oxidants can generate stable post-translational modifications (PTMs) on proteins. Oxidative PTMs are recognised in evolutionarily-conserved innate immune responses. These PTMs can represent neo-epitopes that break tolerance in autoimmune disease. Antibodies targeting these PTMs in diseases e.g. RA and SLE, can be biomarkers.
Collapse
Affiliation(s)
- Brent J. Ryan
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Ahuva Nissim
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Queen Mary, University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Paul G. Winyard
- University of Exeter Medical School, St Luke's Campus, Exeter, Devon EX1 2LU, UK
- Corresponding author.
| |
Collapse
|
48
|
The association among antioxidant enzymes, autoantibodies, and disease severity score in systemic lupus erythematosus: comparison of neuropsychiatric and nonneuropsychiatric groups. BIOMED RESEARCH INTERNATIONAL 2014; 2014:137231. [PMID: 24877055 PMCID: PMC4024413 DOI: 10.1155/2014/137231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 04/01/2014] [Indexed: 11/29/2022]
Abstract
Background. Antioxidative capacity plays an important role in the severity of systemic lupus erythematosus (SLE), which is characterized by autoantibodies. This study aimed to determine the relationship among autoantibody titers, antioxidative stress reserve, and severity of SLE. Methods. The autoantibody titers, clinical markers, antioxidant enzyme levels, and disease activity index (SLEDAI-2k) of 32 SLE patients and 16 healthy controls were compared. We also compared both the neuropsychiatric (NPSLE) and nonneuropsychiatric (non-NPSLE) groups. Results. Superoxide dismutase in red blood cells was significantly lower in the SLE than in the control group. CRP levels are significant higher in SLE patients than in control group (P = 0.034). Among the autoantibodies, anti-U1RNP (P = 0.008), a-Sm (P = 0.027), and anti-ribosomal p (P = 0.028) significantly negatively correlated with glutathione levels. There has no significant correlation between SLE disease activity indexes (SLEDAI) and levels of C3, C4, and antioxidant enzymes. Conclusions. Erythrocyte superoxide dismutase is significantly lower in both NPSLE and non-NPSLE groups. SLE patients have both higher CRP and autoantibodies level and decreased superoxide dismutase level than the healthy control group.
Collapse
|
49
|
Fan X, Wang G, English RD, Firoze Khan M. Proteomic identification of carbonylated proteins in the kidney of trichloroethene-exposed MRL+/+ mice. Toxicol Mech Methods 2013; 24:21-30. [PMID: 24024666 DOI: 10.3109/15376516.2013.843112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Trichloroethene (TCE), a common environmental and occupational pollutant, is associated with multiorgan toxicity. Kidney is one of major target organs affected as a result of TCE exposure. Our previous studies have shown that exposure to TCE causes increased protein oxidation (protein carbonylation) in the kidneys of autoimmune-prone MRL+/+ mice, and suggested a potential role of protein oxidation in TCE-mediated nephrotoxicity. To assess the impact of chronic TCE exposure on protein oxidation, particularly to identify the carbonylated proteins in kidneys, female MRL+/+ mice were treated with TCE at the dose of 2 mg/ml via drinking water for 36 weeks and kidney protein extracts were analyzed for protein carbonyls and carbonylated proteins identified using proteomic approaches (2D gel, Western blot, MALDI TOF/TOF MS/MS, etc.). TCE treatment led to significantly increased protein carbonyls in the kidney protein extracts (20 000 g pellet fraction). Interestingly, among 18 identified carbonylated proteins, 10 were found only in the kidneys of TCE-treated mice, whereas other 8 were present in the kidneys of both control and TCE-treated mice. The identified carbonylated proteins represent skeletal proteins, chaperones, stress proteins, enzymes, plasma protein and proteins involved in signaling pathways. The findings provide a map for further exploring the role of carbonylated proteins in TCE-mediated nephrotoxicity.
Collapse
|
50
|
N-Acetylcysteine protects against trichloroethene-mediated autoimmunity by attenuating oxidative stress. Toxicol Appl Pharmacol 2013; 273:189-95. [PMID: 23993974 DOI: 10.1016/j.taap.2013.08.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 08/09/2013] [Accepted: 08/14/2013] [Indexed: 12/19/2022]
Abstract
Exposure to trichloroethene (TCE), a ubiquitous environmental contaminant, is known to induce autoimmunity both in humans and animal models. However, mechanisms underlying TCE-mediated autoimmunity remain largely unknown. Previous studies from our laboratory in MRL+/+ mice suggest that oxidative stress may contribute to TCE-induced autoimmune response. The current study was undertaken to further assess the role of oxidative stress in TCE-induced autoimmunity by supplementing with an antioxidant N-acetylcysteine (NAC). Groups of female MRL+/+ mice were given TCE, NAC or TCE+NAC for 6 weeks (TCE, 10mmol/kg, i.p., every 4th day; NAC, 250mg/kg/day through drinking water). TCE exposure led to significant increases in serum levels of anti-nuclear, anti-dsDNA and anti-Sm antibodies. TCE exposure also led to significant induction of anti-malondiadelhyde (MDA)- and anti-hydroxynonenal (HNE)-protein adduct antibodies which were associated with increased ANA in the sera along with increased MDA-/HNE-protein adducts in the livers and kidneys, and increases in protein oxidation (carbonylation) in the sera, livers and kidneys, suggesting an overall increase in oxidative stress. Moreover, TCE exposure also resulted in increased release of IL-17 from splenocytes and increases in IL-17 mRNA expression. Remarkably, NAC supplementation attenuated not only the TCE-induced oxidative stress, IL-17 release and mRNA expression, but also the markers of autoimmunity, as evident from decreased levels of ANA, anti-dsDNA and anti-Sm antibodies in the sera. These results provide further support to a role of oxidative stress in TCE-induced autoimmune response. Attenuation of TCE-induced autoimmunity in mice by NAC provides an approach for preventive and/or therapeutic strategies.
Collapse
|