1
|
Mok TC, Mok CC. The Potential Use of Arsenic Trioxide in the Treatment of Systemic Lupus Erythematosus. Int J Mol Sci 2024; 25:9577. [PMID: 39273522 PMCID: PMC11394723 DOI: 10.3390/ijms25179577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Arsenic trioxide (ATO) is now part of the standard regimen for the treatment of newly diagnosed and relapsed acute promyelocytic leukemia. The availability of an oral form of ATO has greatly reduced the incidence of cardiotoxicity as compared to intravenous (IV) administration. Increasing evidence suggests that ATO has anti-inflammatory properties that may be useful for the treatment of autoimmune diseases. These include the modulation of Treg cell activation, Th1/Th2 and Th17/Treg balance, depletion of activated T cells and plasmacytoid dendritic cells, and influence of B-cell differentiation, leading to reduced autoantibody and cytokine production. ATO has also been shown to induce apoptosis of activated fibroblast-like synoviocytes through the generation of reactive oxygen species and alter the gut microbiota in collagen-induced arthritis. Despite the emergence of newer treatment modalities, the treatment of systemic lupus erythematosus (SLE), especially refractory manifestations, remains a challenge, owing to the paucity of effective biological and targeted therapies that are devoid of adverse effects. Oral ATO is an attractive option for the treatment of SLE because of the lower cost of production, convenience of administration, and reduced cardiotoxicity. This article summarizes the anti-inflammatory mechanisms of ATO and its potential application in the treatment of SLE and other rheumatic diseases.
Collapse
Affiliation(s)
- Tsz Ching Mok
- Department of Medicine, Ruttonjee Hospital, Hong Kong SAR, China
| | - Chi Chiu Mok
- Department of Medicine and Geriatrics, Tuen Mun Hospital, Hong Kong SAR, China
| |
Collapse
|
2
|
Yan M, Wang H, Wei R, Li W. Arsenic trioxide: applications, mechanisms of action, toxicity and rescue strategies to date. Arch Pharm Res 2024; 47:249-271. [PMID: 38147202 DOI: 10.1007/s12272-023-01481-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 12/15/2023] [Indexed: 12/27/2023]
Abstract
Arsenical medicine has obtained its status in traditional Chinese medicine for more than 2,000 years. In the 1970s, arsenic trioxide was identified to have high efficacy and potency for the treatment of acute promyelocytic leukemia, which promoted many studies on the therapeutic effects of arsenic trioxide. Currently, arsenic trioxide is widely used to treat acute promyelocytic leukemia and various solid tumors through various mechanisms of action in clinical practice; however, it is accompanied by a series of adverse reactions, especially cardiac toxicity. This review presents a comprehensive overview of arsenic trioxide from preclinical and clinical efficacy, potential mechanisms of action, toxicities, and rescue strategies for toxicities to provide guidance or assistance for the clinical application of arsenic trioxide.
Collapse
Affiliation(s)
- Meng Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China.
| | - Hao Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Rui Wei
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- Pharmacy Department, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenwen Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
3
|
Gill H, Russell N, Kwong YL. Editorial: Acute promyelocytic leukemia - towards a chemotherapy-free approach to cure in all patients, Volume II. Front Oncol 2023; 13:1238486. [PMID: 37441418 PMCID: PMC10335353 DOI: 10.3389/fonc.2023.1238486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 06/15/2023] [Indexed: 07/15/2023] Open
Affiliation(s)
- Harinder Gill
- Department of Medicine, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, the University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Nigel Russell
- Department of Haematology, Nottingham City Hospital and University of Nottingham, Nottingham, United Kingdom
| | - Yok-Lam Kwong
- Department of Medicine, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, the University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
4
|
Chen JZ, Wang LN, Luo XQ, Tang YL. The genomic landscape of sensitivity to arsenic trioxide uncovered by genome-wide CRISPR-Cas9 screening. Front Oncol 2023; 13:1178686. [PMID: 37251921 PMCID: PMC10214836 DOI: 10.3389/fonc.2023.1178686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Introduction Arsenic trioxide (ATO) is a promising anticancer drug for hematological malignancy. Given the dramatic efficacy of acute promyelocytic leukemia (APL), ATO has been utilized in other types of cancers, including solid tumors. Unfortunately, the results were not comparable with the effects on APL, and the resistance mechanism has not been clarified yet. This study intends to identify relevant genes and pathways affecting ATO drug sensitivity through genome-wide CRISPR-Cas9 knockdown screening to provide a panoramic view for further study of ATO targets and improved clinical outcomes. Methods A genome-wide CRISPR-Cas9 knockdown screening system was constructed for ATO screening. The screening results were processed with MAGeCK, and the results were subjected to pathway enrichment analysis using WebGestalt and KOBAS. We also performed protein-protein interaction (PPI) network analysis using String and Cytoscape, followed by expression profiling and survival curve analysis of critical genes. Virtual screening was used to recognize drugs that may interact with the hub gene. Results We applied enrichment analysis and identified vital ATO-related pathways such as metabolism, chemokines and cytokines production and signaling, and immune system responses. In addition, we identified KEAP1 as the top gene relating to ATO resistance. We found that KEAP1 expression was higher in the pan-cancer, including ALL, than in normal tissue. Patients with acute myeloid leukemia (AML) with higher KEAP1 expression had worse overall survival (OS). A virtual screen showed that etoposide and eltrombopag could bind to KEAP1 and potentially interact with ATO. Discussion ATO is a multi-target anticancer drug, and the key pathways regulating its sensitivity include oxidative stress, metabolism, chemokines and cytokines, and the immune system. KEAP1 is the most critical gene regulating ATO drug sensitivity, which is related to AML prognosis and may bind to some clinical drugs leading to an interaction with ATO. These integrated results provided new insights into the pharmacological mechanism of ATO and potentiate for further applications in cancer treatments.
Collapse
Affiliation(s)
- Jun-Zhu Chen
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Li-Na Wang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xue-Qun Luo
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yan-Lai Tang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Chêne C, Rongvaux-Gaïda D, Thomas M, Rieger F, Nicco C, Batteux F. Optimal combination of arsenic trioxide and copper ions to prevent autoimmunity in a murine HOCl-induced model of systemic sclerosis. Front Immunol 2023; 14:1149869. [PMID: 37063915 PMCID: PMC10097895 DOI: 10.3389/fimmu.2023.1149869] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/06/2023] [Indexed: 04/18/2023] Open
Abstract
Introduction Systemic sclerosis (SSc) is a rare chronic autoimmune disease characterized by diffuse fibrosis of the skin and internal organs and vascular abnormalities. The etiology and physiopathology are complex due to the heterogeneity of its overall clinical presentation. Arsenic trioxide (ATO) has been proven to be effective against SSc, sclerodermatous Graft-versus-Host Disease, multiple sclerosis, Crohn's disease or systemic lupus erythematosus animal models and has demonstrated promising effects in human clinical trials. Its efficacy was shown to be related at least in part to the generation of Reactive Oxygen Species (ROS) and the selective deletion of activated immune cells and fibroblasts. However, ATO can induce some adverse effects that must be considered, especially when used for the treatment of a chronic disease. Methods We evaluate here, in vitro and in a mouse model of SSc, the improved efficacy of ATO when associated with a Fenton-like divalent cation, namely copper chloride (CuCl2), also known to trigger the production of ROS. Results In preliminary experiments in vitro, ATO 1 µM + CuCl2 0.5 µM increased ROS production and increased apoptosis of NIH 3T3 murine fibroblasts compared to 1 µM ATO alone. In vivo, in the HOCl-induced mouse model of SSc, co-treatment with ATO 2.5 μg/g + CuCl2 0.5 μg/g significantly alleviated clinical signs such as the thickening of the skin (p<0.01) and cutaneous fibrosis, in a manner equivalent to treatment with ATO 5 µg/g. Our results provide evidence that co-treatment with ATO 2.5 μg/g + CuCl2 0.5 μg/g decreases the number of B cells and the activation of CD4+ T lymphocytes. The co-treatment substantially blocks the NRF2 signaling pathway, increases H2O2 production and results in the improvement of the health status of mice with experimental SSc. Conclusion In conclusion, copper combined with ATO treatment halved the concentration of ATO needed to obtain the same effect as a high dose of ATO alone for the treatment of SSc mice. The strategy of using lower doses of drugs with different mechanisms of action in combination has many potential advantages, the first being to lessen the potential side effects induced by ATO, a drug with side effects quickly increased with dosage.
Collapse
Affiliation(s)
- Charlotte Chêne
- INSERM U1016, Institut Cochin, Département 3I “Infection, Immunité Et Inflammation”, Université Paris Cité, Paris, France
- R&D Department, MEDSENIC SAS, Strasbourg, France
| | | | - Marine Thomas
- INSERM U1016, Institut Cochin, Département 3I “Infection, Immunité Et Inflammation”, Université Paris Cité, Paris, France
| | | | - Carole Nicco
- INSERM U1016, Institut Cochin, Département 3I “Infection, Immunité Et Inflammation”, Université Paris Cité, Paris, France
- *Correspondence: Frédéric Batteux, ; Carole Nicco,
| | - Frédéric Batteux
- INSERM U1016, Institut Cochin, Département 3I “Infection, Immunité Et Inflammation”, Université Paris Cité, Paris, France
- Service d’immunologie Biologique, AP-HP-Centre Université Paris Cité, Hôpital Cochin, Université Paris Cité, Faculté De Médecine, Paris, France
- *Correspondence: Frédéric Batteux, ; Carole Nicco,
| |
Collapse
|
6
|
Kumana CR, Kwong YL, Gill H. Oral arsenic trioxide for treating acute promyelocytic leukaemia: Implications for its worldwide epidemiology and beyond. Front Oncol 2022; 12:1026478. [PMID: 36518307 PMCID: PMC9744132 DOI: 10.3389/fonc.2022.1026478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/31/2022] [Indexed: 07/29/2023] Open
Abstract
This account describes how orally administered Arsenic-trioxide (ATO) therapy influences the epidemiology of acute promyelocytic leukaemia (APL), and how the experience that ensued may expand the indications for oral ATO as a treatment for diseases/disorders other than APL. Over the last two decades, experience with APL patients in Hong Kong treated with an oral regimen comprising ATO, all-trans retinoic acid (ATRA), and ascorbic acid (also known as "AAA") has confirmed a dramatic improvement in overall survival. Over that period, there has been an estimated 60-fold increase in the prevalence of APL (proportion of surviving APL patients in the population on December 31 including those deemed to be 'cured'). In contrast to regimens entailing intravenous (IV) ATO, the consequential therapeutic benefits of using oral ATO have been achieved with much less patient inconvenience and quality of life disruption, reduced burdens on health care facilities (hospitalisations and staff involvement), and much enhanced affordability (retail drug & other cost reductions). Numerous experimental and a few clinical studies suggest that ATO may also have a therapeutic role in many other diseases/disorders. Several such diseases (e.g. autoimmune disorders & idiopathic pulmonary fibrosis) are far more prevalent than APL, which means that very large numbers of patients may potentially benefit from ATO treatment, even if its efficacy is limited to selected populations with these diseases. The known safety of oral ATO and its advantages over repeated long-term IV delivery suggests that this route be used in future clinical studies of its possible role in treating such patients. If the clinical utility of oral ATO treatment is validated for patients enduring any such non-APL diseases, very large numbers of patients may stand to benefit.
Collapse
|
7
|
Mellouk A, Hutteau-Hamel T, Legrand J, Safya H, Benbijja M, Mercier-Nomé F, Benihoud K, Kanellopoulos JM, Bobé P. P2X7 purinergic receptor plays a critical role in maintaining T-cell homeostasis and preventing lupus pathogenesis. Front Immunol 2022; 13:957008. [PMID: 36248812 PMCID: PMC9556828 DOI: 10.3389/fimmu.2022.957008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/30/2022] [Indexed: 11/20/2022] Open
Abstract
The severe lymphoproliferative and lupus diseases developed by MRL/lpr mice depend on interactions between the Faslpr mutation and MRL genetic background. Thus, the Faslpr mutation causes limited disease in C57BL/6 mice. We previously found that accumulating B220+ CD4–CD8– double negative (DN) T cells in MRL/lpr mice show defective P2X7 receptor ( P2X7)-induced cellular functions, suggesting that P2X7 contributes to T-cell homeostasis, along with Fas. Therefore, we generated a B6/lpr mouse strain (called B6/lpr-p2x7KO) carrying homozygous P2X7 knockout alleles. B6/lpr-p2x7KO mice accumulated high numbers of FasL-expressing B220+ DN T cells of CD45RBhighCD44high effector/memory CD8+ T-cell origin and developed severe lupus, characterized by leukocyte infiltration into the tissues, high levels of IgG anti-dsDNA and rheumatoid factor autoantibodies, and marked cytokine network dysregulation. B6/lpr-p2x7KO mice also exhibited a considerably reduced lifespan. P2X7 is therefore a novel regulator of T-cell homeostasis, of which cooperation with Fas is critical to prevent lymphoaccumulation and autoimmunity.
Collapse
Affiliation(s)
- Amine Mellouk
- UMR 996, INSERM, Université Paris-Saclay, Clamart, France
| | | | - Julie Legrand
- Institut André Lwoff, CNRS, Université Paris-Sud, Villejuif, France
| | - Hanaa Safya
- UMR 996, INSERM, Université Paris-Saclay, Clamart, France
| | - Mohcine Benbijja
- Institut André Lwoff, CNRS, Université Paris-Sud, Villejuif, France
| | - Françoise Mercier-Nomé
- UMR 996, INSERM, Université Paris-Saclay, Clamart, France
- Plateforme d’Histologie Immunopathologie de Clamart, IPSIT, INSERM, CNRS, Université Paris-Saclay, Châtenay-Malabry, France
| | - Karim Benihoud
- UMR 9018, Institut Gustave Roussy, CNRS, Université Paris-Saclay, Villejuif, France
| | - Jean M. Kanellopoulos
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Pierre Bobé
- UMR 996, INSERM, Université Paris-Saclay, Clamart, France
- Institut André Lwoff, CNRS, Université Paris-Sud, Villejuif, France
- *Correspondence: Pierre Bobé,
| |
Collapse
|
8
|
The Development and Clinical Applications of Oral Arsenic Trioxide for Acute Promyelocytic Leukaemia and Other Diseases. Pharmaceutics 2022; 14:pharmaceutics14091945. [PMID: 36145693 PMCID: PMC9504237 DOI: 10.3390/pharmaceutics14091945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
Appreciation of the properties of arsenic trioxide (ATO) has redefined the treatment landscape for acute promyelocytic leukaemia (APL) and offers promise as a treatment for numerous other diseases. The benefits of ATO in patients with APL is related to its ability to counteract the effects of PML::RARA, an oncoprotein that is invariably detected in the blood or bone marrow of affected individuals. The PML::RARA oncoprotein is degraded specifically by binding to ATO. Thus ATO, in combination with all-trans retinoic acid, has become the curative treatment for ATO. The multiple mechanisms of action of ATO has also paved the way for application in various condition encompassing autoimmune or inflammatory disorders, solid organ tumours, lymphomas and other subtypes of AML. The development of oral formulation of ATO (oral ATO) has reduced costs of treatment and improved treatment convenience allowing widespread applicability. In this review, we discuss the mechanisms of action of ATO, the development of oral ATO, and the applications of oral ATO in APL and other diseases.
Collapse
|
9
|
Rongvaux-Gaïda D, Dupuis M, Poupon J, Djebrani-Oussedik N, Lemonnier C, Rieger F. High Response Rate and Corticosteroid Sparing with Arsenic Trioxide-Based First-Line Therapy in Chronic Graft-versus-Host Disease after Allogeneic Hematopoietic Stem Cell Transplantation. Transplant Cell Ther 2022; 28:679.e1-679.e11. [DOI: 10.1016/j.jtct.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 07/01/2022] [Accepted: 07/01/2022] [Indexed: 11/16/2022]
|
10
|
Balkrishna A, Solleti SK, Singh H, Singh R, Bhattacharya K, Varshney A. Herbo-metallic ethnomedicine 'Malla Sindoor' ameliorates lung inflammation in murine model of allergic asthma by modulating cytokines status and oxidative stress. JOURNAL OF ETHNOPHARMACOLOGY 2022; 292:115120. [PMID: 35202713 DOI: 10.1016/j.jep.2022.115120] [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: 09/10/2021] [Revised: 02/06/2022] [Accepted: 02/14/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Asthma is the leading inflammatory disease of the airways with inadequate therapeutic options. 'Malla Sindoor' (MS) is a metal-based ethnomedicinal formulation that has been prescribed in the ancient traditional medicinal system for treating chronic inflammations. AIM OF THE STUDY Here, we validated the anti-inflammatory and anti-asthmatic properties of traditional metallic medicine MS in asthmatic mice model and in LPS stimulated human monocytic THP-1 cells, by examining the relevant cellular, biochemical and molecular intermediates. MATERIALS AND METHODS Scanning Electron Microscope (SEM), Electron Dispersive X-ray (EDX), and X-Ray Diffraction (XRD) were performed to characterize MS particles. Allergic asthma was induced in Balb/c mice through intraperitoneal ovalbumin (OVA) injection. Experimental groups include, normal control, disease control, Dexamethasone (2 mg/kg) and three MS treated groups: 4.3 mg/kg, 13 mg/kg, and 39 mg/kg. Quantitative PCR, inflammatory cytokines and anti-oxidant enzymes, and histological analysis were performed, in the treated mice and LPS stimulated human monocytic THP-1 cells for determining the MS efficacy. RESULTS SEM image analysis showed the MS to be heterogenous in shape with a particle size distribution between 100 nm-1 μm. Elemental composition showed the presence of mercury (Hg), arsenic (As), and sulphur (S) along with other elements in the forms of mercury sulfide, arsenic trioxide, and their alloy crystals. OVA-challenge of the Balb/c mice resulted in the development of overt pathological features for allergic asthma including smooth muscle thickening and collagen deposition. Mice receiving MS-exhibited alleviation of allergic asthma features. BAL fluid analysis showed a decrease in the total cell count and decreases in neutrophils, monocytes, lymphocytes, and eosinophils. Further, the stimulated levels of interleukin (IL)-1β, -6, and TNF-α cytokines and antioxidant levels were also reduced upon MS-treatment. At the molecular level, MS-treatment reduced stimulated mRNA expression levels for IL-4, -5, -10, -13, -33, and IFN-γ cytokines. Histological analysis following MS-treatment of OVA-stimulated mice lungs showed a reduction in mucus accumulation in airways, decreases in peribronchial collagen deposition, bronchial smooth muscle thickening, and attenuation of inflammatory cell infiltration. In addition, under in-vitro conditions, MS-treatment attenuated the LPS induced secretion of IL-1β, -6, and TNF-α from THP-1 cells. CONCLUSION Collectively, the results suggest that MS acts as an effective anti-asthmatic and anti-inflammatory agent, by regulating various cellular, biochemical and molecular intermediates.
Collapse
Affiliation(s)
- Acharya Balkrishna
- Drug Discovery and Development Division, Patanjali Research Institute, NH-58, Haridwar, Uttarakhand, India; Department of Allied and Applied Sciences, University of Patanjali, Patanjali Yog Peeth, Roorkee-Haridwar Road, Haridwar, Uttarakhand, India; Patanjali UK Trust, Glasgow, United Kingdom
| | - Siva Kumar Solleti
- Drug Discovery and Development Division, Patanjali Research Institute, NH-58, Haridwar, Uttarakhand, India
| | - Hoshiyar Singh
- Drug Discovery and Development Division, Patanjali Research Institute, NH-58, Haridwar, Uttarakhand, India
| | - Rani Singh
- Drug Discovery and Development Division, Patanjali Research Institute, NH-58, Haridwar, Uttarakhand, India
| | - Kunal Bhattacharya
- Drug Discovery and Development Division, Patanjali Research Institute, NH-58, Haridwar, Uttarakhand, India
| | - Anurag Varshney
- Drug Discovery and Development Division, Patanjali Research Institute, NH-58, Haridwar, Uttarakhand, India; Department of Allied and Applied Sciences, University of Patanjali, Patanjali Yog Peeth, Roorkee-Haridwar Road, Haridwar, Uttarakhand, India; Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, 110 067, India.
| |
Collapse
|
11
|
Quan P, Wang K, Yan S, Wen S, Wei C, Zhang X, Cao J, Yao L. Integrated network analysis identifying potential novel drug candidates and targets for Parkinson's disease. Sci Rep 2021; 11:13154. [PMID: 34162989 PMCID: PMC8222400 DOI: 10.1038/s41598-021-92701-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/14/2021] [Indexed: 11/09/2022] Open
Abstract
This study aimed to identify potential novel drug candidates and targets for Parkinson's disease. First, 970 genes that have been reported to be related to PD were collected from five databases, and functional enrichment analysis of these genes was conducted to investigate their potential mechanisms. Then, we collected drugs and related targets from DrugBank, narrowed the list by proximity scores and Inverted Gene Set Enrichment analysis of drug targets, and identified potential drug candidates for PD treatment. Finally, we compared the expression distribution of the candidate drug-target genes between the PD group and the control group in the public dataset with the largest sample size (GSE99039) in Gene Expression Omnibus. Ten drugs with an FDR < 0.1 and their corresponding targets were identified. Some target genes of the ten drugs significantly overlapped with PD-related genes or already known therapeutic targets for PD. Nine differentially expressed drug-target genes with p < 0.05 were screened. This work will facilitate further research into the possible efficacy of new drugs for PD and will provide valuable clues for drug design.
Collapse
Affiliation(s)
- Pusheng Quan
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
| | - Kai Wang
- Center of TOF-PET/CT/MR, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
| | - Shi Yan
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
| | - Shirong Wen
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
| | - Chengqun Wei
- Department of General Practice, Heilongjiang Provincial Hospital, Harbin, 150081, China
| | - Xinyu Zhang
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
| | - Jingwei Cao
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, 150081, China
| | - Lifen Yao
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
12
|
Li C, Chu T, Zhang Z, Zhang Y. Single Cell RNA-Seq Analysis Identifies Differentially Expressed Genes of Treg Cell in Early Treatment-Naive Rheumatoid Arthritis By Arsenic Trioxide. Front Pharmacol 2021; 12:656124. [PMID: 34108876 PMCID: PMC8181733 DOI: 10.3389/fphar.2021.656124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/28/2021] [Indexed: 12/27/2022] Open
Abstract
Objective: Early treatment-naïve rheumatoid arthritis (RA) has defective regulatory T (Treg) cells and increased inflammation response. In this study, we aim to illustrate the regulation of Treg cells in pathogenesis of early rheumatoid arthritis by arsenic trioxide (As2O3). Methods: We studied the effects of As2O3 on gene expression in early treatment-naïve RA Treg cells with single cell RNA-seq (scRNA-seq). Treg cells were sorted from peripheral blood mononuclear cells (PBMCs) and purified by fluorescence-activated cell sorting (FACS) and cultured with or without As2O3 (at 0.1 µM) for 24 h. Total RNA was isolated and sequenced, and functional analysis was performed against the Gene Ontology (GO) database. Results for selected genes were confirmed with RT-qPCR. Results: As2O3 exerts no significant effect on CD4+ T-cell apoptosis under physical condition, and selectively modulate CD4+ T cells toward Treg cells not Th17 cells under special polarizing stimulators. As2O3 increased the expression of 200 and reduced that of 272 genes with fold change (FC) 2.0 or greater. Several genes associated with inflammation, Treg-cell activation and differentiation as well as glucose and amino acids metabolism were among the most strongly affected genes. GO function analysis identified top ten ranked significant biological process (BPs), molecular functions (MFs), and cell components (CCs) in treatment and nontreatment Treg cells. In GO analysis, genes involved in the immunoregulation, cell apoptosis and cycle, inflammation, and cellular metabolism were enriched among the significantly affected genes. The KEGG pathway enrichment analysis identified the forkhead box O (FoxO) signal pathway, apoptosis, cytokine–cytokine receptor interaction, cell cycle, nuclear factor-kappa B (NF-κB) signaling pathway, tumor necrosis factor α (TNF-α), p53 signaling pathway, and phosphatidylinositol 3′-kinase (PI3K)-Akt signaling pathway were involved in the pathogenesis of early treatment-naïve RA. Conclusion: This is the first study investigating the genome-wide effects of As2O3 on the gene expression of treatment-naïve Treg cells. In addition to clear anti-inflammatory and immunoregulation effects, As2O3 affect amino acids and glucose metabolism in Treg cells, an observation that might be particularly important in the metabolic phenotype of treatment-naïve RA.
Collapse
Affiliation(s)
- Chunling Li
- Department of Rheumatology and Immunology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China.,Department of Rheumatology and Immunology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tianshu Chu
- Department of Rheumatology and Immunology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhiyi Zhang
- Department of Rheumatology and Immunology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yue Zhang
- Department of Rheumatology and Immunology, First Affiliated Hospital of Harbin Medical University, Harbin, China.,Department of Rheumatology, Immunology and Gerontology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, China.,Shenzhen Futian Hospital of Rheumatic Diseases, Shenzhen, China
| |
Collapse
|
13
|
Hamidou M, Néel A, Poupon J, Amoura Z, Ebbo M, Sibilia J, Viallard JF, Gaborit B, Volteau C, Hardouin JB, Hachulla E, Rieger F. Safety and efficacy of low-dose intravenous arsenic trioxide in systemic lupus erythematosus: an open-label phase IIa trial (Lupsenic). Arthritis Res Ther 2021; 23:70. [PMID: 33658052 PMCID: PMC7927234 DOI: 10.1186/s13075-021-02454-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Lupus animal model has shown that arsenic trioxide (ATO), a treatment of acute promyelocytic leukaemia, could be effective in SLE. This is the first clinical study to determine the safety and efficacy of a short course of intravenous ATO in patients with active SLE. METHODS This phase IIa, open-label, dose-escalating study enrolled 11 adult SLE patients with a non-organ threatening disease, clinically active despite conventional therapy. Patients received 10 IV infusions of ATO within 24 days. The first group received 0.10 mg/kg per injection, with dose-escalating to 0.15 mg/kg in a second group, and to 0.20 mg/kg in a third group. The primary endpoint was the occurrence of adverse events (AEs) and secondary endpoints were the number of SLE Responder Index 4 (SRI-4) responders at week 24 and reduction of corticosteroid dosage. In an exploratory analysis, we collected long-term data for safety and attainment of lupus low disease activity state (LLDAS). RESULTS Four serious AEs occurred (grade 3 neutropenia, osteitis, neuropathy), 2 of which were attributable to ATO (neutropenia in the 2 patients treated with mycophenolate). Two patients suffered a severe flare during the last 4 weeks of the trial. At W24, five patients among 10 were SRI-4 responders. Overall, mean corticosteroid dosage decreased from 11.25 mg/day at baseline to 6 mg/day at W24 (P < 0.01). In the long term, 6 patients attained LLDAS at W52, which continued at last follow-up (median LLDAS duration 3 years, range 2-4). CONCLUSIONS A short course of ATO has an acceptable safety profile in SLE patients and encouraging efficacy. TRIAL REGISTRATION ClinicalTrials.gov, NCT01738360 registered 30 November 2012.
Collapse
Affiliation(s)
- Mohamed Hamidou
- Department of Internal Medicine, CHU Nantes, Nantes Université, Nantes, France.
| | - Antoine Néel
- Department of Internal Medicine, CHU Nantes, Nantes Université, Nantes, France
| | - Joel Poupon
- Department of Biological Toxicology, AP-HP, Lariboisière Hospital, University Paris VII, Paris, France
| | - Zahir Amoura
- Department of Internal Medicine 2, Centre National de Référence pour le Lupus, Institut E3M, Hôpital Pitié-Salpétrière, Paris, France
| | - Mikael Ebbo
- Service de Médecine Interne, Aix Marseille Univ, APHM, CNRS, INSERM, CIML, Hôpital de la Timone, Marseille, France
| | - Jean Sibilia
- Department of Rheumatology, University of Strasbourg, Strasbourg, France
| | | | - Benjamin Gaborit
- Department of Internal Medicine, CHU Nantes, Nantes Université, Nantes, France
| | - Christelle Volteau
- Plateforme de Méthodologie et Biostatistiques, CHU Nantes, Université de Nantes, Nantes, France
| | | | - Eric Hachulla
- Department of Internal Medicine, Centre de Référence des Maladies Autoimmunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), University of Lille, Lille, France
| | - François Rieger
- MEDSENIC, SAS, a company with CNRS participation, Strasbourg, France
| |
Collapse
|
14
|
Mihaylova N, Bradyanova S, Chipinski P, Chausheva S, Kyurkchiev D, Tchorbanov AI. Monoclonal antibody therapy that targets phospholipid-binding protein delays lupus activity in MRL/lpr mice. Scand J Immunol 2020; 92:e12915. [PMID: 32533866 DOI: 10.1111/sji.12915] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/14/2020] [Accepted: 06/03/2020] [Indexed: 11/28/2022]
Abstract
Systemic lupus erythematosus is an autoimmune syndrome characterized by the development of autoantibodies to a wide range of antigens. Together with B cells, respective self-reactive T cells have an important contribution in disease progression as being responsible for inflammatory cytokines secretion, B cell activation and promoting amplification of the autoimmune response. Annexin A1 is expressed by many cell types and binds to phospholipids in a Ca2+ -dependent manner. Abnormal expression of annexin A1 was found on activated B and T cells in both murine and human autoimmunity suggesting its potential role as a therapeutic target. In the present study, we have investigated the possibility to suppress autoimmune manifestation in spontaneous mouse model of lupus using anti-annexin A1 antibody. Groups of lupus-prone MRL/lpr mice were treated with the anti-annexin A1 monoclonal antibody, and the disease activity and survival of the animals were following up. Flow cytometry, ELISA assays, and histological and immunofluorescence kidney analyses were used to determine the levels of Annexin A1 expression, cytokines, anti-dsDNA antibodies and kidney injuries. The administration of this monoclonal antibody to MRL/lpr mice resulted in suppression of IgG anti-dsDNA antibody production, modulated IL-10 secretion, decreased disease activity and prolonged survival compared with the control group.
Collapse
Affiliation(s)
- Nikolina Mihaylova
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Silviya Bradyanova
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Petroslav Chipinski
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Stela Chausheva
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Dobroslav Kyurkchiev
- Laboratory of Clinical Immunology, University Hospital 'Sv.I.Rilski', Medical University Sofia, Sofia, Bulgaria
| | - Andrey I Tchorbanov
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria.,National Institute of Immunology, Sofia, Bulgaria
| |
Collapse
|
15
|
Ye Y, Ricard L, Siblany L, Stocker N, De Vassoigne F, Brissot E, Lamarthée B, Mekinian A, Mohty M, Gaugler B, Malard F. Arsenic trioxide induces regulatory functions of plasmacytoid dendritic cells through interferon- α inhibition. Acta Pharm Sin B 2020; 10:1061-1072. [PMID: 32642412 PMCID: PMC7332672 DOI: 10.1016/j.apsb.2020.01.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 12/04/2019] [Accepted: 12/16/2019] [Indexed: 12/29/2022] Open
Abstract
Arsenic trioxide (As2O3) is recently found to have therapeutic potential in systemic sclerosis (SSc), a life-threatening multi-system fibrosing autoimmune disease with type I interferon (IFN-I) signature. Chronically activated plasmacytoid dendritic cells (pDCs) are responsible for IFN-I secretion and are closely related with fibrosis establishment in SSc. In this study, we showed that high concentrations of As2O3 induced apoptosis of pDCs via mitochondrial pathway with increased BAX/BCL-2 ratio, while independent of reactive oxygen species generation. Notably, at clinical relevant concentrations, As2O3 preferentially inhibited IFN-α secretion as compared to other cytokines such as TNF-α, probably due to potent down-regulation of the total protein and mRNA expression, as well as phosphorylation of the interferon regulatory factor 7 (IRF7). In addition, As2O3 induced a suppressive phenotype, and in combination with cytokine inhibition, it down-regulated pDCs’ capacity to induce CD4+ T cell proliferation, Th1/Th22 polarization, and B cell differentiation towards plasmablasts. Moreover, chronically activated pDCs from SSc patients were not resistant to the selective IFN-α inhibition, and regulatory phenotype induced by As2O3. Collectively, our data suggest that As2O3 could target pDCs and exert its treatment efficacy in SSc, and more autoimmune disorders with IFN-I signature.
Collapse
|
16
|
Ye Y, Gaugler B, Mohty M, Malard F. Plasmacytoid dendritic cell biology and its role in immune-mediated diseases. Clin Transl Immunology 2020; 9:e1139. [PMID: 32489664 PMCID: PMC7248678 DOI: 10.1002/cti2.1139] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 04/27/2020] [Accepted: 04/27/2020] [Indexed: 12/26/2022] Open
Abstract
Plasmacytoid dendritic cells (pDCs) are a unique subset of dendritic cells specialised in secreting high levels of type I interferons. pDCs play a crucial role in antiviral immunity and have been implicated in the initiation and development of many autoimmune and inflammatory diseases. This review summarises the latest advances in recent years in several aspects of pDC biology, with special focus on pDC heterogeneity, pDC development via the lymphoid pathway, and newly identified proteins/pathways involved in pDC trafficking, nucleic acid sensing and interferon production. Finally, we also highlight the current understanding of pDC involvement in autoimmunity and alloreactivity, and opportunities for pDC‐targeting therapies in these diseases. These new insights have contributed to answers to several fundamental questions remaining in pDC biology and may pave the way to successful pDC‐targeting therapy in the future.
Collapse
Affiliation(s)
- Yishan Ye
- INSERM, Centre de Recherche Saint-Antoine (CRSA) Sorbonne Université Paris France.,Bone Marrow Transplantation Center The First Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
| | - Béatrice Gaugler
- INSERM, Centre de Recherche Saint-Antoine (CRSA) Sorbonne Université Paris France
| | - Mohamad Mohty
- INSERM, Centre de Recherche Saint-Antoine (CRSA) Sorbonne Université Paris France.,Service d'Hématologie Clinique et Thérapie Cellulaire AP-HP, Hôpital Saint-Antoine Sorbonne Université Paris France
| | - Florent Malard
- INSERM, Centre de Recherche Saint-Antoine (CRSA) Sorbonne Université Paris France.,Service d'Hématologie Clinique et Thérapie Cellulaire AP-HP, Hôpital Saint-Antoine Sorbonne Université Paris France
| |
Collapse
|
17
|
An K, Xue MJ, Zhong JY, Yu SN, Lan TS, Qi ZQ, Xia JJ. Arsenic trioxide ameliorates experimental autoimmune encephalomyelitis in C57BL/6 mice by inducing CD4 + T cell apoptosis. J Neuroinflammation 2020; 17:147. [PMID: 32375831 PMCID: PMC7201567 DOI: 10.1186/s12974-020-01829-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is an immune-mediated disease of the central nervous system characterized by severe white matter demyelination. Because of its complex pathogenesis, there is no definite cure for MS. Experimental autoimmune encephalomyelitis (EAE) is an ideal animal model for the study of MS. Arsenic trioxide (ATO) is an ancient Chinese medicine used for its therapeutic properties with several autoimmune diseases. It is also used to inhibit acute immune rejection due to its anti-inflammatory and immunosuppressive properties. However, it is unclear whether ATO has a therapeutic effect on EAE, and the underlying mechanisms have not yet been clearly elucidated. In this study, we attempted to assess whether ATO could be used to ameliorate EAE in mice. METHODS ATO (0.5 mg/kg/day) was administered intraperitoneally to EAE mice 10 days post-immunization for 8 days. On day 22 post-immunization, the spinal cord, spleen, and blood were collected to analyze demyelination, inflammation, microglia activation, and the proportion of CD4+ T cells. In vitro, for mechanistic studies, CD4+ T cells were sorted from the spleen of naïve C57BL/6 mice and treated with ATO and then used for an apoptosis assay, JC-1 staining, imaging under a transmission electron microscope, and western blotting. RESULTS ATO delayed the onset of EAE and alleviated the severity of EAE in mice. Treatment with ATO also attenuated demyelination, alleviated inflammation, reduced microglia activation, and decreased the expression levels of IL-2, IFN-γ, IL-1β, IL-6, and TNF-α in EAE mice. Moreover, the number and proportion of CD4+ T cells in the spinal cord, spleen, and peripheral blood were reduced in ATO-treated EAE mice. Finally, ATO induced CD4+ T cell apoptosis via the mitochondrial pathway both in vitro and in vivo. Additionally, the administration of ATO had no adverse effect on the heart, liver, or kidney function, nor did it induce apoptosis in the spinal cord. CONCLUSIONS Overall, our findings indicated that ATO plays a protective role in the initiation and progression of EAE and has the potential to be a novel drug in the treatment of MS.
Collapse
Affiliation(s)
- Ke An
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Meng-Jiao Xue
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jia-Ying Zhong
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Sheng-Nan Yu
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, China
| | - Tian-Shu Lan
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen Medical College, Xiamen, Fujian, China
| | - Zhong-Quan Qi
- School of Medicine, Guangxi University, Nanning, Guangxi, China.
| | - Jun-Jie Xia
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
18
|
Ye Y, Gaugler B, Mohty M, Malard F. Old dog, new trick: Trivalent arsenic as an immunomodulatory drug. Br J Pharmacol 2020; 177:2199-2214. [PMID: 32022256 DOI: 10.1111/bph.15011] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/19/2019] [Accepted: 01/28/2020] [Indexed: 12/19/2022] Open
Abstract
Trivalent arsenic (As(III)) is recently found to be an immunomodulatory agent. As(III) has therapeutic potential in several autoimmune and inflammatory diseases in vivo. In vitro, it selectively induces apoptosis of immune cells due to different sensitivity. At a non-toxic level, As(III) shows its multifaceted nature by inducing either pro- or anti-inflammatory functions of immune subsets. These effects are exerted by either As(III)-protein interactions or as a consequence of As(III)-induced homeostasis imbalance. The immunomodulatory properties also show synergistic effects of As(III) with cancer immunotherapy. In this review, we summarize the immunomodulatory effects of As(III), focusing on the effects of As(III) on immune subsets in vitro, on mouse models of immune-related diseases, and the role of As(III) in cancer immunotherapy. Updates of the mechanisms of action, the pioneer clinical trials, dosing, and adverse events of therapeutic As(III) are also provided.
Collapse
Affiliation(s)
- Yishan Ye
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Béatrice Gaugler
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, Paris, France
| | - Mohamad Mohty
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, Paris, France
| | - Florent Malard
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, Paris, France
| |
Collapse
|
19
|
Tian W, Wang Z, Tang NN, Li JT, Liu Y, Chu WF, Yang BF. Ascorbic Acid Sensitizes Colorectal Carcinoma to the Cytotoxicity of Arsenic Trioxide via Promoting Reactive Oxygen Species-Dependent Apoptosis and Pyroptosis. Front Pharmacol 2020; 11:123. [PMID: 32153415 PMCID: PMC7047232 DOI: 10.3389/fphar.2020.00123] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 01/29/2020] [Indexed: 12/22/2022] Open
Abstract
Arsenic trioxide (ATO) is an effective therapeutic agent against acute promyelocytic leukemia (APL); however, its anti-tumor effect on solid tumors such as colorectal cancer (CRC) is still in debate. Ascorbic acid (AA) also produces a selective cytotoxic activity against tumor cells. Here, we exploit the potential benefit of ATO/AA combination in generating cytotoxicity to CRC cells, which may lay the groundwork for the potential combinational chemotherapy of CRCs. According to the results, we found that ATO and AA effectively inhibited the viability of human CRC cells in a synergistic manner. AA and ATO corporately activated caspase-3 to trigger apoptosis and upregulated the expression of caspase-1 and promoted formation of inflammasomes to induce pyroptosis. Furthermore, the stimulation of reactive oxygen species (ROS) overproduction was demonstrated as a subcellular mechanism for apoptosis and pyroptosis induced by ATO/AA combination treatment. Our findings suggest that ATO combination with a conventional dosage of AA offers an advantage for killing CRC cells. The synergistic action of ATO/AA combination might be considered a plausible strategy for the treatment of CRC and perhaps other solid tumors as well.
Collapse
Affiliation(s)
- Wei Tian
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang, China
| | - Zhuo Wang
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang, China
| | - Nan-Nan Tang
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang, China
| | - Jia-Tong Li
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang, China
| | - Yu Liu
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang, China
| | - Wen-Feng Chu
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang, China
| | - Bao-Feng Yang
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University at Harbin, Heilongjiang, China
| |
Collapse
|
20
|
Zhao Y, Mu Z, Cai L, Liu X, Jia J, Zhang J. Tetra-arsenic tetra-sulfide ameliorates lupus syndromes by inhibiting IL-17 producing double negative T cells. Dermatol Ther 2019; 32:e12849. [PMID: 30707471 DOI: 10.1111/dth.12849] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 11/26/2018] [Accepted: 01/24/2019] [Indexed: 12/24/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease of uncertain etiology that affects multiple tissues and organs. Tetra-arsenic tetra-sulfide (As4 S4 ), a traditional Chinese medicine, is effective on acute promyelocytic leukemia with mild side effects. In our previous study, BXSB lupus-prone mice treated with As4 S4 has showed improved monocytosis, decreased serum interleukin (IL)-6 and suppressed skin, liver and renal lesions with well-tolerance. In this study, we explored the effect and mechanism of As4 S4 on the MRL/lpr mice. MRL/lpr and wild MRL/MpJ mice were divided into control and As4 S4 treatment groups and dosed with As4 S4 or placebo for 8 weeks. We found that As4 S4 prevented the skin, renal and lung lesions of MRL/lpr mice. As4 S4 significantly decreased the double negative T (DN T) cells and reduced the serum levels of IL-17, IL-10, and antinuclear antibodies titer. Further results revealed that the FasL was decreased, and activated caspases elevated in DN T cells in As4 S4 treated MRL/lpr mice. Taken together, As4 S4 could selectively suppresses DN T cells by inducing apoptosis. It also reduced inflammatory cytokines IL-17, which may be produced by DN T cells. As4 S4 may represent a new therapy for SLE.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Dermatology, People's Hospital of Peking University, Beijing, China
| | - Zhanglei Mu
- Department of Dermatology, People's Hospital of Peking University, Beijing, China
| | - Lin Cai
- Department of Dermatology, People's Hospital of Peking University, Beijing, China
| | - Xiaojing Liu
- Department of Dermatology, People's Hospital of Peking University, Beijing, China
| | - Jun Jia
- Department of Dermatology, People's Hospital of Peking University, Beijing, China
| | - Jianzhong Zhang
- Department of Dermatology, People's Hospital of Peking University, Beijing, China
| |
Collapse
|
21
|
Abstract
In 1992, arsenic trioxide (As2O3, ATO) was demonstrated to be an effective therapeutic agent against acute promyelocytic leukemia (APL), rekindling attention to ATO applications in U.S. Food and Drug Administration clinical trials for the treatment of cancers, such as leukemia, lymphomas, and solid tumors. ATO is a potent chemotherapeutic drug that can also be used to treat other diseases, such as autoimmune diseases, because it affects multiple pathways including apoptosis induction, differentiation stimulation, and proliferation inhibition. As inflammation is a critical component of disease progression, ATO is a feasible treatment option based on its ability to protect against inflammation. However, ATO is also a well-known carcinogen because of its pro-inflammatory effect. This review will focus on the double-sided effects of ATO on inflammation as well as the relevant mechanisms underlying these effects, aiming to provide a rational understanding of how ATO effects the immune system. We especially aim to provide a comprehensive overview of our current knowledge of how ATO influences inflammation.
Collapse
|
22
|
Bonam SR, Wang F, Muller S. Autophagy: A new concept in autoimmunity regulation and a novel therapeutic option. J Autoimmun 2018; 94:16-32. [PMID: 30219390 DOI: 10.1016/j.jaut.2018.08.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 08/27/2018] [Accepted: 08/28/2018] [Indexed: 02/07/2023]
Abstract
Nowadays, pharmacologic treatments of autoinflammatory diseases are largely palliative rather than curative. Most of them result in non-specific immunosuppression, which can be associated with broad disruption of natural and induced immunity with significant and sometimes serious unwanted injuries. Among the novel strategies that are under development, tools that modulate the immune system to restore normal tolerance mechanisms are central. In these approaches, peptide therapeutics constitute a class of agents that display many physicochemical advantages. Within this class of potent drugs, the phosphopeptide P140 is very promising for treating patients with lupus, and likely also patients with other chronic inflammatory diseases. We discovered that P140 targets autophagy, a finely orchestrated catabolic process, involved in the regulation of inflammation and in the biology of immune cells. In vitro, P140 acts directly on a particular form of autophagy called chaperone-mediated autophagy, which seems to be hyperactivated in certain subsets of lymphocytes in lupus and in other autoinflammatory settings. In lupus, the "correcting" effect of P140 on autophagy results in a weaker signaling of autoreactive T cells, leading to a significant improvement of pathophysiological status of treated mice. These findings also demonstrated ex vivo in human cells, open novel avenues of therapeutic intervention in pathological conditions, in which specific and not general targeting is highly pursued in the context of the new action plans for personalized medicines.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- CNRS-University of Strasbourg, Biotechnology and Cell Signaling, Illkirch, France; CNRS-University of Strasbourg, Laboratory of Excellence Medalis, France
| | - Fengjuan Wang
- CNRS-University of Strasbourg, Biotechnology and Cell Signaling, Illkirch, France; CNRS-University of Strasbourg, Laboratory of Excellence Medalis, France
| | - Sylviane Muller
- CNRS-University of Strasbourg, Biotechnology and Cell Signaling, Illkirch, France; CNRS-University of Strasbourg, Laboratory of Excellence Medalis, France; University of Strasbourg Institute for Advanced Study, Strasbourg, France.
| |
Collapse
|
23
|
Ahn H, Kim J, Kang SG, Yoon SI, Ko HJ, Kim PH, Hong EJ, An BS, Lee E, Lee GS. Mercury and arsenic attenuate canonical and non-canonical NLRP3 inflammasome activation. Sci Rep 2018; 8:13659. [PMID: 30209319 PMCID: PMC6135747 DOI: 10.1038/s41598-018-31717-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 08/22/2018] [Indexed: 11/09/2022] Open
Abstract
Exposure to heavy metals can cause several diseases associated with the immune system. Although the effects of heavy metals on production of inflammatory cytokines have been previously studied, the role of heavy metals in inflammasome activation remains poorly studied. The inflammasome is an intracellular multi-protein complex that detects intracellular danger signals, resulting in inflammatory responses such as cytokine maturation and pyroptosis. In this study, we elucidated the effects of four heavy metals, including cadmium (Cd), mercury (Hg), arsenic (As), and lead (Pb), on the activation of NLRP3, NLRC4, and AIM2 inflammasomes. In our results, mercury and arsenic inhibited interleukin (IL)-1β and IL-18 secretion resulting from canonical and non-canonical NLRP3 inflammasome activation in macrophages and attenuated elevation of serum IL-1β in response to LPS treatment in mice. In the mechanical studies, mercury interrupted production of mitochondrial reactive oxygen species, release of mitochondrial DNA, and activity of recombinant caspase-1, whereas arsenic down-regulated expression of promyelocytic leukemia protein. Both mercury and arsenic inhibited Asc pyroptosome formation and gasdermin D cleavage. Thus, we suggest that exposure to mercury and/or arsenic could disrupt inflammasome-mediated inflammatory responses, which might cause unexpected side effects.
Collapse
Affiliation(s)
- Huijeong Ahn
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Jeongeun Kim
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Seung Goo Kang
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Sung-Il Yoon
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Hyun-Jeong Ko
- Laboratory of Microbiology and Immunology, College of Pharmacy, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Pyeung-Hyeun Kim
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Eui-Ju Hong
- College of Veterinary Medicine and Institute of Veterinary Science, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Beum-Soo An
- Department of Biomaterial Science, College of Natural Resources and Life Science, Pusan National University, Gyeongsangnam-do, 50612, Republic of Korea
| | - Eunsong Lee
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Geun-Shik Lee
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea.
| |
Collapse
|
24
|
Hu H, Chen E, Li Y, Zhu X, Zhang T, Zhu X. Effects of Arsenic Trioxide on INF-gamma Gene Expression in MRL/lpr Mice and Human Lupus. Biol Trace Elem Res 2018; 184:391-397. [PMID: 29159556 DOI: 10.1007/s12011-017-1206-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 11/15/2017] [Indexed: 02/02/2023]
Abstract
Arsenic trioxide (As2O3; ATO), a traditional Chinese medicine, is used to treat patients with acute promye-locytic leukemia, while its application for treatment of systemic lupus erythematosus (SLE) is still under evaluation. The high expression of INF-gamma (INF-γ) is a primary pathogenic factor in SLE. It is found that ATO can reduce INF-γ expression levels in lupus-prone mice, whereas it is not clear whether ATO has the same effect on SLE patients. Therefore, this study was to investigate the underlying mechanism of the effects of ATO on the expression of INF-γ in splenocytes of MRL/lpr mice and PBMCs of human lupus. The mRNA and protein expression levels of INF-γ were assessed by real-time RT-PCR and ELISA, respectively. The histone acetylation status of the INF-γ promoter and the binding of RNA polymerase II (RNA Pol II) to the INF-γ promoter were detected using a chromatin immunoprecipitation (ChIP) technique. The mRNA and protein expression levels of INF-γ decreased in both splenocytes of MRL/lpr mice and PBMCs of SLE patients with ATO treatment, which were accompanied by reduced histone H4 and H3 acetylation in INF-γ promoter and decreased combination of RNA Pol II to the INF-γ promoter. Therefore, ATO may reduce the expression level of the INF-γ by altering the levels of INF-γ promoter acetylation and the combination of RNA Pol II to the INF-γ promoter in splenocytes of MRL/lpr mice and PBMCs of SLE patients.
Collapse
Affiliation(s)
- Hongye Hu
- Department of Surgical Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Enjiu Chen
- Department of Pneumology, The People Hospital of Pingyang, Wenzhou, Zhejiang Province, China
| | - Yongji Li
- Department of Rheumatology, The First Affiliated Hospital of Wenzhou Medical University, Nan Bai Xiang Street, Ouhai District, Wenzhou, Zhejiang Province, 325000, China
| | - Xiaochun Zhu
- Department of Rheumatology, The First Affiliated Hospital of Wenzhou Medical University, Nan Bai Xiang Street, Ouhai District, Wenzhou, Zhejiang Province, 325000, China
| | - Ting Zhang
- Department of Rheumatology, The First Affiliated Hospital of Wenzhou Medical University, Nan Bai Xiang Street, Ouhai District, Wenzhou, Zhejiang Province, 325000, China
| | - Xiaofang Zhu
- Department of Rheumatology, The First Affiliated Hospital of Wenzhou Medical University, Nan Bai Xiang Street, Ouhai District, Wenzhou, Zhejiang Province, 325000, China.
| |
Collapse
|
25
|
|
26
|
Zaalberg A, Moradi Tuchayi S, Ameri AH, Ngo KH, Cunningham TJ, Eliane JP, Livneh M, Horn TD, Rosman IS, Musiek A, Anadkat MJ, Demehri S. Chronic Inflammation Promotes Skin Carcinogenesis in Cancer-Prone Discoid Lupus Erythematosus. J Invest Dermatol 2018; 139:62-70. [PMID: 30030152 DOI: 10.1016/j.jid.2018.06.185] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 05/20/2018] [Accepted: 06/24/2018] [Indexed: 11/17/2022]
Abstract
High-risk skin cancer is a rare, but severe, complication associated with discoid lupus erythematosus (DLE). Chronic scar, inflammation, UVR, and immunosuppressive medications are proposed explanations for this heightened skin cancer risk; however, the exact mechanism driving skin carcinogenesis in DLE is unknown. The distinct co-localization of multiple independent skin cancers with areas of active inflammation in two DLE patients followed over 8 years strongly suggested that lupus inflammation promotes skin carcinogenesis in DLE. To investigate this clinical observation, we subjected lupus-prone MRL/lpr and control (MRL/n) mice to a skin carcinogenesis protocol. Skin tumors developed preferentially within the cutaneous lupus inflammation without scarring in MRL/lpr mice (P < 0.01). The inflammation in MRL/lpr skin was characterized by the accumulation of regulatory T cells, mast cells, M2 macrophages, and markedly elevated transforming growth factor-β1 and IL-6 levels, which have been linked to tumor promotion. Tacrolimus treatment reduced skin inflammation and blocked cancer development in MRL/lpr mice (P = 0.0195). A similar tumor-promoting immune environment was detected in SCCs and the perilesional skin of cancer-prone DLE patients. Therefore, discoid lupus inflammation promotes skin cancer in high-risk DLE patients, and blocking the inflammation may be critical for preventing this life-threatening complication of DLE.
Collapse
Affiliation(s)
- Anniek Zaalberg
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sara Moradi Tuchayi
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Amir H Ameri
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kenneth H Ngo
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Trevor J Cunningham
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jean-Pierre Eliane
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Maia Livneh
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas D Horn
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ilana S Rosman
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA; Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Amy Musiek
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Milan J Anadkat
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Shadmehr Demehri
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
27
|
Komai T, Inoue M, Okamura T, Morita K, Iwasaki Y, Sumitomo S, Shoda H, Yamamoto K, Fujio K. Transforming Growth Factor-β and Interleukin-10 Synergistically Regulate Humoral Immunity via Modulating Metabolic Signals. Front Immunol 2018; 9:1364. [PMID: 29963056 PMCID: PMC6010538 DOI: 10.3389/fimmu.2018.01364] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 06/01/2018] [Indexed: 12/13/2022] Open
Abstract
Inhibitory cytokines, such as transforming growth factor-β (TGF-β) and interleukin-10 (IL-10), are humoral factors involved in the suppressive function of regulatory T cells and play critical roles in maintaining immune homeostasis. However, TGF-β and IL-10 also have pleiotropic effects and induce humoral immune responses depending on conditions, and thus their therapeutic application to autoimmune diseases remains limited. Here, we show that a combination of TGF-β and IL-10, but not single cytokine, is required to suppress B cell activation induced by toll-like receptor (TLR) stimulation. In in vivo analyses, the simultaneous presence of TGF-β and IL-10 effectively suppressed TLR-mediated antigen-specific immune responses and ameliorated pathologies in imiquimod (TLR7 agonist)-induced lupus model and lupus-prone MRL/lpr mice. Intriguingly, TGF-β and IL-10 synergistically modulated transcriptional programs and suppressed cellular energetics of both glycolysis and oxidative phosphorylation via inhibition of the mammalian target of rapamycin complex 1 (mTORC1)/S6 kinase 1 (S6K1) pathway in TLR-stimulated B cells. On the other hand, enhancement of mTOR signaling and mitochondrial biosynthesis in TLR-stimulated B cells counteracted the synergistic inhibitory effects. The inhibitory cytokine synergy of TGF-β and IL-10 via suppression of energy metabolism was also observed in human TLR-stimulated B cells. There is increasing evidence supporting the importance of adequate metabolic signals in various immune cells to exert their immune function. In this study, we have shown that a previously unrecognized synergy of inhibitory cytokines regulates systemic humoral immune responses via modulating immunometabolism in B cells. Our findings indicate that inhibition of B cell metabolism mediated by two synergistic cytokines contributes to the induction of immune tolerance and could be a new therapeutic strategy for autoimmune diseases such as systemic lupus erythematosus.
Collapse
Affiliation(s)
- Toshihiko Komai
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mariko Inoue
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Max Planck-The University of Tokyo Center for Integrative Inflammology, The University of Tokyo, Tokyo, Japan
| | - Kaoru Morita
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yukiko Iwasaki
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shuji Sumitomo
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hirofumi Shoda
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Yamamoto
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Max Planck-The University of Tokyo Center for Integrative Inflammology, The University of Tokyo, Tokyo, Japan
- Laboratory for Autoimmune Diseases, Center for Integrative Medical Sciences, RIKEN, Kanagawa, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
28
|
Ikeda T, Fujii H, Nose M, Kamogawa Y, Shirai T, Shirota Y, Ishii T, Harigae H. Bortezomib treatment induces a higher mortality rate in lupus model mice with a higher disease activity. Arthritis Res Ther 2017; 19:187. [PMID: 28800777 PMCID: PMC5553803 DOI: 10.1186/s13075-017-1397-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 07/27/2017] [Indexed: 01/06/2023] Open
Abstract
Background Bortezomib (Bz) is a proteasome inhibitor that directly targets antibody-producing plasma cells. We recently reported the first randomized control trial that evaluated the effects of Bz in patients with systemic lupus erythematosus (SLE). In that study, we demonstrated that Bz treatment is associated with many adverse reactions in patients with refractory disease. In the present study, we examine the therapeutic and toxic effects of Bz on MRL/MpJ-lpr/lpr (MRL/lpr) mice with severe disease activity. Methods Female MRL/lpr mice at 10 and 14 weeks of age were treated with phosphate buffered saline (PBS) (n = 19), Bz (750 μg/kg twice weekly) (n = 27), or cyclophosphamide (Cyc) (1 mg/body, once in 2 weeks) (n = 20). Cellular subsets, serum immunoglobulin, anti-double-stranded DNA (anti-dsDNA) antibody titer, and a pathological index of glomerulonephritis were then analyzed at 22 weeks of age. Survival curves of the 10-week-old and 14-week-old Bz-treated groups were compared. Blood counts, creatinine, liver enzymes, and serum cytokine levels were measured 1 week after Bz treatment. Gene expression profiling of spleens from Bz and Cyc treatment mice were compared with those from control mice. Results The anti-dsDNA antibody levels were significantly higher in 14-week-old than in 10-week-old mice, indicating a higher disease activity at 14 weeks. A significant decrease in the number of splenic cells and glomerulonephritis index was observed in Bz-treated and Cyc-treated mice. Bz, but not Cyc, significantly decreased serum immunoglobulin and anti-dsDNA antibody titer levels. Survival curve analysis revealed a significantly higher mortality rate in 14-week-old than in 10-week-old Bz-treated and control groups. Following two injections of Bz, serum IL-6 and TNF-α levels were significantly more elevated in 14-week-old than in 10-week-old mice. Potentially immunogenic molecules, such as heat shock proteins, were characteristically upregulated in spleens of Bz-treated but not Cyc-treated mice. Conclusions In spite of its therapeutic effect, Bz treatment had more toxic effects associated with increased proinflammatory cytokine levels in mice with a higher disease activity. Understanding the mechanism of the toxicity and developing preventive strategies against it is important for the safe clinical application of Bz in human SLE. Electronic supplementary material The online version of this article (doi:10.1186/s13075-017-1397-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tomoko Ikeda
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Hiroshi Fujii
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan.
| | - Masato Nose
- Institue for Promotion of Advanced Science and Technology, Ehime University, Matsuyama, Japan
| | - Yukiko Kamogawa
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Tsuyoshi Shirai
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Yuko Shirota
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Tomonori Ishii
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Hideo Harigae
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| |
Collapse
|
29
|
Zhang J, Li C, Zheng Y, Lin Z, Zhang Y, Zhang Z. Inhibition of angiogenesis by arsenic trioxide via TSP-1-TGF-β1-CTGF-VEGF functional module in rheumatoid arthritis. Oncotarget 2017; 8:73529-73546. [PMID: 29088724 PMCID: PMC5650279 DOI: 10.18632/oncotarget.19867] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 07/11/2017] [Indexed: 01/25/2023] Open
Abstract
Angiogenesis is a critical factor for rheumatoid arthritis (RA). Although anti-TNF biologics work effectively on some RA patients, concerns have been raised about the possible increased development of malignancies alongside such treatments. Arsenic trioxide (As2O3) has attracted worldwide attention and has been reported to treat some cancers. However, the effects of As2O3 on angiogenesis in the RA synovium remain unclear. Here, we report a systematic increased expression of TSP-1, TGF-β1, CTGF and VEGF in supernatants of a RA fibroblast-like synoviocytes (RA-FLS) and human dermal microvascular endothelial cells (HDMECs) co-culture compared with those from a normal human fibroblast-like synoviocytes (NH-FLS) and HDMECs co-culture. This increased expression may up-regulate endothelial tube formation and transwell migration, as well as microvessel sprouting in ex vivo aortic ring assay. These networked angiogenic factors mainly form a functional module regulating angiogenesis in the RA synovium. We show that As2O3 inhibits angiogenesis in the collagen-induced arthritis (CIA) synovium and consequently arthritis severity via significant suppression of TSP-1, TGF-β1, CTGF and VEGF expression in the CIA synovium, plus in the RA-FLS and HDMECs co-culture as well as NH-FLS and HDMECs co-culture system along with the presence or absence of TNF-α treatment. Thus As2O3 has a significant anti-angiogenesis effect on the RA-FLS and CIA synovium via its inhibition of the RA angiogenic functional module of TSP-1, TGF-β1, CTGF and VEGF and may have a potential for treating RA beyond cancer therapy.
Collapse
Affiliation(s)
- Juan Zhang
- Department of Rheumatology, The First Affiliated Hospital, Harbin Medical University, Nan Gang, Harbin, China
| | - Chunling Li
- Department of Rheumatology, The First Affiliated Hospital, Harbin Medical University, Nan Gang, Harbin, China
| | - Yining Zheng
- Department of Rheumatology, The First Affiliated Hospital, Harbin Medical University, Nan Gang, Harbin, China
| | - Zhiguo Lin
- Department of Rheumatology, The First Affiliated Hospital, Harbin Medical University, Nan Gang, Harbin, China
| | - Yue Zhang
- Department of Rheumatology, The First Affiliated Hospital, Harbin Medical University, Nan Gang, Harbin, China
| | - Zhiyi Zhang
- Department of Rheumatology, The First Affiliated Hospital, Harbin Medical University, Nan Gang, Harbin, China
| |
Collapse
|
30
|
Abstract
The genomic revolution in the past decade fuelled by breathtaking advances in sequencing technologies has defined several new genetic diseases of the immune system. Many of these newly characterized diseases are a result of defects in genes involved in immune regulation. The discovery of these diseases has opened a vista of new therapeutic possibilities. Immunomodulatory agents, a hitherto unexplored therapeutic option in primary immunodeficiency diseases have been tried in a host of these newly described maladies. These agents have been shown conclusively to favorably modulate immune responses, resulting in abatement of clinical manifestations both in experimental models and patients. While some of the treatment options have been approved for therapeutic use or have been shown to be of merit in open-label trials, others have been shown to be efficacious in a handful of clinical cases, animal models, and cell lines. Interferon γ is approved for use in chronic granulomatous disease (CGD) to reduce the burden of infection and and has a good long-term efficacy. Recombinant human IL7 therapy has been shown increase the peripheral CD4 and CD8 T cell counts in patients with idiopathic CD4. Anti-IL1 agents are approved for the management of cryopyrin-related autoinflammatory syndrome, and their therapeutic efficacy is being increasingly recognized in other autoinflammatory syndromes and CGD. Mammalian target of rapamycin (mTOR) inhibitors have been proven useful in autoimmune lymphoproliferative syndrome (ALPS) and in IPEX syndrome. Therapies reported to be potential use in case reports include abatacept in CTLA4 haploinsufficiency and LRBA deficiency, ruxolitinib in gain-of-function STAT1, tocilizumab in gain-of-function STAT3 defect, mTOR inhibitors in PIK3CD activation, magnesium in XMEN syndrome, and pioglitazone in CGD. Treatment options of merit in human cell lines include interferon α and interferon β in TLR3 and UNC-93B deficiencies, anti-interferon therapy in SAVI, and Rho-kinase inhibitors in TTC7A deficiency. Anti-IL17 agents have show efficacy in animal models of leukocyte adhesion defect (LAD) and ALPS. This topical review explores the use of various immunomodulators and other biological agents in the context of primary immunodeficiency and autoinflammatory diseases.
Collapse
|
31
|
Mihaylova N, Bradyanova S, Chipinski P, Herbáth M, Chausheva S, Kyurkchiev D, Prechl J, Tchorbanov AI. Annexin A1 as a target for managing murine pristane-induced systemic lupus erythematosus. Autoimmunity 2017; 50:257-268. [DOI: 10.1080/08916934.2017.1300884] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Nikolina Mihaylova
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Silviya Bradyanova
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Petroslav Chipinski
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Melinda Herbáth
- MTA-ELTE Immunology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Stela Chausheva
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Dobroslav Kyurkchiev
- Laboratory of Clinical Immunology, University Hospital ‘St.I.Rilski’, Medical University Sofia, Sofia, Bulgaria
| | - József Prechl
- MTA-ELTE Immunology Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Andrey I. Tchorbanov
- Laboratory of Experimental Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
32
|
Li P, Huang P, Yang Y, Hao M, Peng H, Li F. Updated Understanding of Autoimmune Lymphoproliferative Syndrome (ALPS). Clin Rev Allergy Immunol 2016; 50:55-63. [PMID: 25663566 DOI: 10.1007/s12016-015-8466-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Autoimmune lymphoproliferative syndrome (ALPS), a disorder characterized by immune dysregulation due to disrupted lymphocyte homeostasis, is mainly resulted from the mutations in FAS-mediated apoptotic pathway. In addition, other mutations of the genes such as Fas-ligand (FASLG), Caspase 10 (CASP10) and Caspase 8 (CASP8), NRAS and KRAS have also been observed in a small number of patients with ALPS or ALPS-related disorders. However, approximately 20-30% of patients with ALPS have unidentified defect. Its clinical manifestations observed in multiple family members include unexplained lymphadenopathy, hepatosplenomegaly, autoimmune cytopenias such as thrombocytopenia, neutropenia, and anemia due to excessive production of antibodies by lymphocytes, elevated number of double-negative T (DNT) cells, and increased risk of lymphoma. As a very rare disease, ALPS was first characterized in the early 1990s. More than 300 families with hereditary ALPS have been reported till now; nearly 500 patients from these families have been studied and followed worldwide over the last 20 years. ALPS has historically considered as a primary immune defect presenting in early childhood, however, recent studies have shown that it may be more common than previous thought because adult onset presentation is increasingly becoming recognized and more adult ALPS patients are diagnosed. The new genetic and biological insights have improved the understanding of ALPS and a number of targeted therapeutic strategies such as mycophenolate mofetil, sirolimus, and pentostatin have been successfully applied in ALPS patients with promising treatment efficacy. This article comprehensively reviews the clinical and laboratory manifestations, new research advances in the molecular pathogenesis, diagnosis and treatments of this disorder.
Collapse
Affiliation(s)
- Pu Li
- Department of Hematology, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Ping Huang
- State Drug Clinical Trial Agency, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Ye Yang
- Department of Internal Medicine, College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Mu Hao
- Department of Internal Medicine, College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Disease Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Hongwei Peng
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zheng Street, Nanchang, Jiangxi, 330006, China
| | - Fei Li
- Department of Hematology, The First Affiliated Hospital of Nanchang University, 17 Yongwai Zheng Street, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
33
|
Prednisone treatment inhibits the differentiation of B lymphocytes into plasma cells in MRL/MpSlac-lpr mice. Acta Pharmacol Sin 2015; 36:1367-76. [PMID: 26456588 DOI: 10.1038/aps.2015.76] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Accepted: 06/11/2015] [Indexed: 01/04/2023] Open
Abstract
AIM A number of evidence shows that the differentiation of B lymphocytes into plasma cells plays an important role in lupus pathogenesis. In this study we investigated how prednisone, a classical therapeutic drug for autoimmune diseases, regulated plasma cell differentiation in MRL/MpSlac-lpr mice. METHODS MRL/lpr mice were treated with prednisone (2.5 or 5 mg·kg(-1)·d(-1), ig) for 13 weeks, and the proteinuria levels and survival times were monitored. After the mice were euthanized, blood sample, spleen and thymus were collected. The serum levels of anti-dsDNA antibody, anti-nuclear antibody, IL-21, and IL-10 were detected using ELISA kits. Subsets of splenic B and T lymphocytes were quantified with flow cytometry. Transcription factor Blimp-1 and Bcl-6 expression was determined using qPCR and Western blot. RESULTS Prednisone treatment dose-dependently attenuated the lupus symptoms in MRL/lpr mice with decreased proteinuria levels, prolonged survival times, decreased serum anti-nuclear antibody levels, and reduced spleen and thymus indices. Prednisone treatment also significantly decreased the elevated percentages of plasma cells and plasma cell precursors, decreased the percentages of activated T cells, and increased the frequency of CD4(+)CD62L(+) cells, demonstrated that decreased anti-nuclear antibodies and improvements in lupus symptoms were associated with decreased plasma cells. Furthermore, prednisone treatment decreased serum IL-21 and IL-10 levels and reduced the expression of splenic Blimp-1 and Bcl-6 (two key regulatory factors for plasma cell differentiation) in MRL/lpr mice. CONCLUSION Prednisone treatment restricts B lymphocyte differentiation into plasma cells in MRL/lpr mice, which may be correlated with the inhibition of IL-21 production and the restoration of the balance between Blimp-1 and Bcl-6.
Collapse
|
34
|
Liu SH, Yang RS, Yen YP, Chiu CY, Tsai KS, Lan KC. Low-Concentration Arsenic Trioxide Inhibits Skeletal Myoblast Cell Proliferation via a Reactive Oxygen Species-Independent Pathway. PLoS One 2015; 10:e0137907. [PMID: 26359868 PMCID: PMC4567280 DOI: 10.1371/journal.pone.0137907] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 08/22/2015] [Indexed: 12/25/2022] Open
Abstract
Myoblast proliferation and differentiation are essential for skeletal muscle regeneration. Myoblast proliferation is a critical step in the growth and maintenance of skeletal muscle. The precise action of inorganic arsenic on myoblast growth has not been investigated. Here, we investigated the in vitro effect of inorganic arsenic trioxide (As2O3) on the growth of C2C12 myoblasts. As2O3 decreased myoblast growth at submicromolar concentrations (0.25–1 μM) after 72 h of treatment. Submicromolar concentrations of As2O3 did not induce the myoblast apoptosis. Low-concentration As2O3 (0.5 and 1 μM) significantly suppressed the myoblast cell proliferative activity, which was accompanied by a small proportion of bromodeoxyuridine (BrdU) incorporation and decreased proliferating cell nuclear antigen (PCNA) protein expression. As2O3 (0.5 and 1 μM) increased the intracellular arsenic content but did not affect the reactive oxygen species (ROS) levels in the myoblasts. Cell cycle analysis indicated that low-concentrations of As2O3 inhibited cell proliferation via cell cycle arrest in the G1 and G2/M phases. As2O3 also decreased the protein expressions of cyclin D1, cyclin E, cyclin B1, cyclin-dependent kinase (CDK) 2, and CDK4, but did not affect the protein expressions of p21 and p27. Furthermore, As2O3 inhibited the phosphorylation of Akt. Insulin-like growth factor-1 significantly reversed the inhibitory effect of As2O3 on Akt phosphorylation and cell proliferation in the myoblasts. These results suggest that submicromolar concentrations of As2O3 alter cell cycle progression and reduce myoblast proliferation, at least in part, through a ROS-independent Akt inhibition pathway.
Collapse
Affiliation(s)
- Shing Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Rong-Sen Yang
- Departments of Orthopaedic, College of Medicine and Hospital, National Taiwan University, Taipei, Taiwan
| | - Yuan-Peng Yen
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chen-Yuan Chiu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Keh-Sung Tsai
- Departments of Laboratory Medicine, College of Medicine and Hospital, National Taiwan University, Taipei, Taiwan
| | - Kuo-Cheng Lan
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
35
|
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
|
36
|
Zhang YF, Zhang M, Huang XL, Fu YJ, Jiang YH, Bao LL, Maimaitiyiming Y, Zhang GJ, Wang QQ, Naranmandura H. The combination of arsenic and cryptotanshinone induces apoptosis through induction of endoplasmic reticulum stress-reactive oxygen species in breast cancer cells. Metallomics 2014; 7:165-73. [PMID: 25412289 DOI: 10.1039/c4mt00263f] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Arsenic trioxide has been successfully used for the treatment of patients with acute promyelocytic leukemia (APL) worldwide. Recently, it has also been further developed to treat solid tumors in clinical trials. However, the therapeutic effects on malignant tumors appeared to be unsatisfactory, as these cells exhibited resistance towards arsenic. In this study, we explored new therapeutic strategies for treatment of human breast cancer MCF-7 cells based on arsenic metabolites. The MCF-7 cells were exposed to three arsenic species, namely, inorganic arsenite (iAs(III)) and its intermediate metabolites monomethylarsonous acid (MMA(III)) and dimethylarsinous acid (DMA(III)) either alone or in combination with cryptotanshinone (CPT) to establish their anticancer effects against MCF-7 cells. Surprisingly, MCF-7 cells were shown to be resistant to both iAs(III) and CPT when used alone; however, they were shown to be relatively sensitive to treatment when exposed to MMA(III) and DMA(III) alone. Conversely, the combination of MMA(III) with CPT showed significantly enhanced anticancer effects on MCF-7 cells at low doses, but no appreciable effect was observed upon exposure to the other two arsenic species with CPT. In addition, remarkable redistribution of pro-apoptosis related proteins Bax and Bak was observed in the mitochondria, together with activation of poly(ADP-ribose) polymerase (PARP) and caspase-9 after exposure to the combination of MMA(III) with CPT. Furthermore, we clearly found that induction of apoptosis in MCF-7 cells was predominantly triggered by endoplasmic reticulum (ER) stress after exposure to the combination of MMA(III) with CPT.
Collapse
Affiliation(s)
- Yan Fang Zhang
- Department of Toxicology, School of Medicine and Public Health, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Benbijja M, Mellouk A, Bobé P. Sensitivity of leukemic T-cell lines to arsenic trioxide cytotoxicity is dependent on the induction of phosphatase B220/CD45R expression at the cell surface. Mol Cancer 2014; 13:251. [PMID: 25410152 PMCID: PMC4252024 DOI: 10.1186/1476-4598-13-251] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 11/06/2014] [Indexed: 11/16/2022] Open
Abstract
Background Arsenic trioxide (As2O3) is highly effective in treating acute promyelocytic leukemia (APL), but shows more variable therapeutic efficacy for other types of hematological malignancies. Previously, we reported that As2O3 selectively eliminates pathogenic B220-expressing T cells in autoimmune MRL/lpr mice. We investigated herein the relationship between As2O3 sensitivity of leukemic T-cell lines and the expression levels of the B220 isoform of transmembrane tyrosine phosphatase CD45. Methods GSH content, O2- production, and B220, HSP70, Fas and FasL membrane expression was measured by flow cytometry. Subcellular localization of B220 was determined by imaging flow cytometry. Cell death was analyzed by morphological changes, annexin V and propidium iodide staining, and caspase 8 and 9 activation. B220 mRNA expression was analyzed by RT-PCR. Activated NF-κB p50 was quantified by a DNA binding ELISA. Results We selected human (Jurkat, Jurkat variant J45.01, HPB-ALL) and mouse (EL-4, BW5147, L1210) T-cell lines for their marked differences in As2O3 sensitivity over a large range of doses (1 to 20 μM). Differences in redox status cannot explain the dramatic differences in As2O3 sensitivity observed among the T-cell lines. Unexpectedly, we found that B220 is differentially induced on As2O3-treated T-cell lines. As2O3 treatment for 24 h induced low (HPB-ALL), intermediate (Jurkat) and high (EL-4, BW5147) levels of B220 membrane expression, membrane-bound HSP70 and cell death, but inhibited NF-κB p50 nuclear translocation. When high levels of B220 expression were achieved with low doses of As2O3, the T-cell lines died by apoptosis only. When high doses of As2O3 were required to induce B220 expression, leukemic T cells died by both apoptosis and necrosis. Conclusions Cellular redox status is not essential for As2O3 sensitivity of leukemic T cells, suggesting the existence of additional factors determining their sensitivity to As2O3 cytotoxicity. Phosphatase B220 could be such a factor of sensitivity. As2O3 treatment inhibits NF-κB p50 nuclear translocation, and induces B220 expression and cell death in a dose and time dependent manner. The levels of B220 induction on leukemic T cells strictly correlate with both the extent and form of cell death, B220 might therefore play a checkpoint role in death pathways. Electronic supplementary material The online version of this article (doi:10.1186/1476-4598-13-251) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | - Pierre Bobé
- Institut Jacques Monod, CNRS, Université Paris Diderot, Paris, France.
| |
Collapse
|
38
|
Watanabe R, Fujii H, Shirai T, Saito S, Ishii T, Harigae H. Autophagy plays a protective role as an anti-oxidant system in human T cells and represents a novel strategy for induction of T-cell apoptosis. Eur J Immunol 2014; 44:2508-20. [DOI: 10.1002/eji.201344248] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 03/24/2014] [Accepted: 04/30/2014] [Indexed: 12/14/2022]
Affiliation(s)
- Ryu Watanabe
- Department of Hematology and Rheumatology; Tohoku University Graduate School of Medicine; Sendai Japan
| | - Hiroshi Fujii
- Department of Hematology and Rheumatology; Tohoku University Graduate School of Medicine; Sendai Japan
| | - Tsuyoshi Shirai
- Department of Hematology and Rheumatology; Tohoku University Graduate School of Medicine; Sendai Japan
| | - Shinichiro Saito
- Department of Hematology and Rheumatology; Tohoku University Graduate School of Medicine; Sendai Japan
| | - Tomonori Ishii
- Department of Hematology and Rheumatology; Tohoku University Graduate School of Medicine; Sendai Japan
| | - Hideo Harigae
- Department of Hematology and Rheumatology; Tohoku University Graduate School of Medicine; Sendai Japan
| |
Collapse
|
39
|
Gesheva V, Kerekov N, Nikolova K, Mihaylova N, Todorov T, Nikolova M, Tchorbanov A. Suppression of dsDNA-specific B lymphocytes reduces disease symptoms in SCID model of mouse lupus. Autoimmunity 2014; 47:162-72. [PMID: 24502777 DOI: 10.3109/08916934.2014.883502] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Self-specific B cells play a main role in the pathogenesis of lupus. This autoimmune disease is characterized by the generation of autoantibodies against self antigens, and the elimination of B and T cells involved in the pathological immune response is a logical approach for effective therapy. We have previously constructed a chimeric molecule by coupling a DNA-mimotope peptides to an anti-CD32 antibody. Using this protein molecule for the treatment of lupus-prone MRL/lpr mice, we suppressed selectively the autoreactive B-lymphocytes by cross-linking B cell receptors with the inhibitory FcγRIIb receptors. This approach was limited by the development of anti-chimeric antibodies in MRL mice. In order to avoid this problem, we established a murine severe combined immunodeficiency lupus model, allowing a long-term chimera therapy. Elimination of the double-stranded DNA-specific B cells by chimera therapy in MRL-transferred immunodeficient mice resulted in inhibition of T cell proliferation and prevented the appearance of IgG anti-DNA antibodies and of proteinuria.
Collapse
Affiliation(s)
- Vera Gesheva
- Department of Immunology, Institute of Microbiology, Bulgarian Academy of Sciences , Sofia , Bulgaria
| | | | | | | | | | | | | |
Collapse
|
40
|
Liu P, Xu S, Zhang M, Wang WW, Zhang YF, Rehman K, Naranmandura H, Chen Z. Anticancer activity in human multiple myeloma U266 cells: synergy between cryptotanshinone and arsenic trioxide. Metallomics 2014; 5:871-8. [PMID: 23529539 DOI: 10.1039/c3mt20272k] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Arsenic trioxide (As2O3) has been recently established as one of the most effective drugs for the treatment of patients with acute promyelocytic leukemia. However, it has exhibited to be less efficient for the non-promyelocytic leukaemia or other types of malignant tumors. The purpose of the present study was to explore new therapeutic strategies based on As2O3 for human multiple myeloma. Here, we first report cryptotanshinone (CPT) and As2O3 synergy for enhanced cytotoxicity in human multiple myeloma U266 cells. In particular, the apoptosis related proteins (e.g., cleaved poly (ADP-ribose) polymerase (PARP), caspase-3 and -9) were significantly increased by the combination treatment (iAs(III) + CPT), whereas, the expression of survival proteins such as Bcl-2 and survivin was suppressed, suggesting that the induction of apoptosis through mitochondrial-mediated apoptotic pathway. In addition, there were no appreciable effects observed in cells after exposure to either As2O3 or CPT alone. In order to better understand the molecular mechanism, we further determined the phosphorylation of STAT3, JNK, ERK and p38. Interestingly, phosphorylation of JNK and p38 were remarkably induced by combination treatment, and no significant inhibition of STAT3 or ERK was observed. In addition, induction of apoptosis in human multiple myeloma cells was partially abrogated only by pretreatment with JNK inhibitor and not by p38 inhibitor, suggesting that JNK pathway may play an important role in induction of apoptosis by the combination of iAs(III) and CPT. Further studies are needed to evaluate this synergistic anticancer effect in vivo. In the near future, this new approach might be used clinically for multiple myeloma (MM) treatment.
Collapse
Affiliation(s)
- Pei Liu
- Zhejiang Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Maier NK, Crown D, Liu J, Leppla SH, Moayeri M. Arsenic trioxide and other arsenical compounds inhibit the NLRP1, NLRP3, and NAIP5/NLRC4 inflammasomes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 192:763-70. [PMID: 24337744 PMCID: PMC3884817 DOI: 10.4049/jimmunol.1301434] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Inflammasomes are large cytoplasmic multiprotein complexes that activate caspase-1 in response to diverse intracellular danger signals. Inflammasome components termed nucleotide-binding oligomerization domain-like receptor (NLR) proteins act as sensors for pathogen-associated molecular patterns, stress, or danger stimuli. We discovered that arsenicals, including arsenic trioxide and sodium arsenite, inhibited activation of the NLRP1, NLRP3, and NAIP5/NLRC4 inflammasomes by their respective activating signals, anthrax lethal toxin, nigericin, and flagellin. These compounds prevented the autoproteolytic activation of caspase-1 and the processing and secretion of IL-1β from macrophages. Inhibition was independent of protein synthesis induction, proteasome-mediated protein breakdown, or kinase signaling pathways. Arsenic trioxide and sodium arsenite did not directly modify or inhibit the activity of preactivated recombinant caspase-1. Rather, they induced a cellular state inhibitory to both the autoproteolytic and substrate cleavage activities of caspase-1, which was reversed by the reactive oxygen species scavenger N-acetylcysteine but not by reducing agents or NO pathway inhibitors. Arsenicals provided protection against NLRP1-dependent anthrax lethal toxin-mediated cell death and prevented NLRP3-dependent neutrophil recruitment in a monosodium urate crystal inflammatory murine peritonitis model. These findings suggest a novel role in inhibition of the innate immune response for arsenical compounds that have been used as therapeutics for a few hundred years.
Collapse
Affiliation(s)
- Nolan K. Maier
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Devorah Crown
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jie Liu
- Center for Molecular Medicine, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stephen H. Leppla
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mahtab Moayeri
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
42
|
Zhao Y, Wen G, Qiao Z, Xu H, Sun Q, Huang H, Shan S, Mu Z, Zhang J. Effects of tetra-arsenic tetra-sulfide on BXSB lupus-prone mice: a pilot study. Lupus 2014; 22:469-76. [PMID: 23554035 DOI: 10.1177/0961203313478302] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease of uncertain etiology that affects multiple tissues and organs. Arsenic trioxide (ATO) has been used in lupus-prone mice with a regulatory effect on immune abnormality. Tetra-arsenic tetra-sulfide (As4S4), a traditional Chinese medicine, is effective on acute promyelocytic leukemia with mild side effects than ATO. In this study, a pilot study was performed to investigate the effects and the mechanism of As4S4 on the lupus-prone BXSB mice. Improvement of monocytosis (p<0.05) in spleen and decreased serum interleukin-6 (IL-6) (p=0.0277) were observed with As4S4 treatment. As4S4-treated mice exhibited amelioration of skin, liver and renal disease with mild side effects. Histological analysis revealed that As4S4 suppressed immune complex deposition, mesangial proliferation and inflammatory cell infiltration in kidney and liver. Our study support that As4S4 selectively suppresses cutaneous lupus and nephritis in BXSB mice and might be a potential treatment for SLE.
Collapse
Affiliation(s)
- Y Zhao
- Department of Dermatology, Peking University People's Hospital, China
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Multiple cysteine residues are necessary for sorting and transport activity of the arsenite permease Acr3p from Saccharomyces cerevisiae. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:747-55. [PMID: 24291645 DOI: 10.1016/j.bbamem.2013.11.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 11/13/2013] [Accepted: 11/20/2013] [Indexed: 11/21/2022]
Abstract
The yeast transporter Acr3p is a low affinity As(III)/H(+) and Sb(III)/H(+) antiporter located in the plasma membrane. It has been shown for bacterial Acr3 proteins that just a single cysteine residue, which is located in the middle of the fourth transmembrane region and conserved in all members of the Acr3 family, is essential for As(III) transport activity. Here, we report a systematic mutational analysis of all nine cysteine residues present in the Saccharomyces cerevisiae Acr3p. We found that mutagenesis of highly conserved Cys151 resulted in a complete loss of metalloid transport function. In addition, lack of Cys90 and Cys169, which are conserved in eukaryotic members of Acr3 family, impaired Acr3p trafficking to the plasma membrane and greatly reduced As(III) efflux, respectively. Mutagenesis of five other cysteines in Acr3p resulted in moderate reduction of As(III) transport capacities and sorting perturbations. Our data suggest that interaction of As(III) with multiple thiol groups in the yeast Acr3p may facilitate As(III) translocation across the plasma membrane.
Collapse
|
44
|
Selective inhibition of the NLRP3 inflammasome by targeting to promyelocytic leukemia protein in mouse and human. Blood 2013; 121:3185-94. [DOI: 10.1182/blood-2012-05-432104] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Key Points
PML selectively activates NLRP3 inflammasome. Targeting to PML could be used to attenuate NLRP3 inflammasome–associated pathogenesis.
Collapse
|
45
|
Li K, Zhang L, Xiang X, Gong S, Ma L, Xu L, Wang G, Liu Y, Ji X, Liu S, Chen P, Zeng H, Li J. Arsenic trioxide alleviates airway hyperresponsiveness and promotes apoptosis of CD4+ T lymphocytes: evidence for involvement of the ER stress-CHOP pathway. Ir J Med Sci 2013; 182:573-83. [PMID: 23494705 DOI: 10.1007/s11845-013-0928-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 02/15/2013] [Indexed: 11/29/2022]
Abstract
BACKGROUND Asthma is a chronic inflammatory disorder of the airway. Arsenic trioxide (ATO) is an ancient Chinese medicine, which is used to treat psoriasis, asthma, and acute promyelocytic leukemia. AIM We wanted to research the effect of arsenic trioxide on asthma. METHODS Using a murine model of asthma, the airway hyperresponsiveness was conducted by the Buxco pulmonary function apparatus. Total cell counts of BALF were counted with a counting chamber. Histopathological analysis of lung tissues was conducted by hematoxylin-eosin or periodic acid-schiff stain. CD4+T cells were purified from the spleen by positive selection, using immunomagnetic beads. Apoptosis measurements were done with Annexin-V/PI staining. Western blot analysis and real time-PCR were performed to assess the expression of C/EBP-homologous protein (CHOP) and glucose-regulated protein 78 (GRP78), respectively. RNA interference was conducted to inhibit the expression of CHOP. RESULTS We found that arsenic trioxide treatment alleviated airway hyperresponsiveness and reduced inflammation of the lung in asthmatic mice. Furthermore, arsenic trioxide treatment promoted apoptosis of CD4+T cells in vivo and in vitro. When CD4+T cells were cultured with arsenic trioxide for 5 h at a concentration of 5 μM, the expression of GRP78 and CHOP was increased. Treatment of CD4+T cells with CHOP siRNA, provided partial resistance to arsenic trioxide-induced apoptosis of CD4+T cells CONCLUSIONS These data demonstrated that arsenic trioxide can reduce the severity of asthma attacks and induce the apoptosis of CD4+ T cell which the ER stress-CHOP pathway involved.
Collapse
Affiliation(s)
- K Li
- Department of Respiratory Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Wahl DR, Byersdorfer CA, Ferrara JLM, Opipari AW, Glick GD. Distinct metabolic programs in activated T cells: opportunities for selective immunomodulation. Immunol Rev 2013; 249:104-15. [PMID: 22889218 DOI: 10.1111/j.1600-065x.2012.01148.x] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
For several decades, it has been known that T-cell activation in vitro leads to increased glycolytic metabolism that fuels proliferation and effector function. Recently, this simple model has been complicated by the observation that different T-cell subsets differentially regulate fundamental metabolic pathways under the control of distinct molecular regulators. Although the majority of these data have been generated in vitro, several recent studies have documented the metabolism of T cells activated in vivo. Here, we review the recent data surrounding the differential regulation of metabolism by distinct T-cell subsets in vitro and in vivo and discuss how differential metabolic regulation might facilitate T-cell function vis-à-vis proliferation, survival, and energy production. We further discuss the important therapeutic implications of differential metabolism across T-cell subsets and review recent successes in exploiting lymphocyte metabolism to treat immune-mediated diseases.
Collapse
Affiliation(s)
- Daniel R Wahl
- Chemical Biology Doctoral Program, University of Michigan, Ann Arbor, MI 48109-1055, USA
| | | | | | | | | |
Collapse
|
47
|
Kavian N, Marut W, Servettaz A, Nicco C, Chéreau C, Lemaréchal H, Borderie D, Dupin N, Weill B, Batteux F. Reactive oxygen species-mediated killing of activated fibroblasts by arsenic trioxide ameliorates fibrosis in a murine model of systemic sclerosis. ACTA ACUST UNITED AC 2013; 64:3430-40. [PMID: 22576901 DOI: 10.1002/art.34534] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE In patients with systemic sclerosis (SSc), activated fibroblasts produce reactive oxygen species (ROS) that stimulate their proliferation and collagen synthesis. By analogy with tumor cells that undergo apoptosis upon cytotoxic treatment that increases ROS levels beyond a lethal threshold, we tested whether activated fibroblasts could be selectively killed by the cytotoxic molecule arsenic trioxide (As(2) O(3) ) in a murine model of SSc. METHODS SSc was induced in BALB/c mice by daily intradermal injections of HOCl. Mice were simultaneously treated with daily intraperitoneal injections of As(2) O(3) . RESULTS As(2) O(3) limited dermal thickness and inhibited collagen deposition, as assessed by histologic examination and measurement of mouse skin and lung collagen contents. As(2) O(3) abrogated vascular damage, as shown by serum vascular cell adhesion molecule 1 level, and inhibited the production of autoantibodies, interleukin-4 (IL-4), and IL-13 by activated T cells. These beneficial effects were mediated through ROS generation that selectively killed activated fibroblasts containing low levels of glutathione. CONCLUSION Our findings indicate that treatment with As(2) O(3) dramatically improves skin and lung fibrosis in a mouse model of SSc, providing a rationale for the evaluation of As(2) O(3) treatment in patients with SSc.
Collapse
Affiliation(s)
- Niloufar Kavian
- Université Paris Descartes, EA 1833, and Hôpital Cochin, AP-HP, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Le Gall SM, Legrand J, Benbijja M, Safya H, Benihoud K, Kanellopoulos JM, Bobé P. Loss of P2X7 receptor plasma membrane expression and function in pathogenic B220+ double-negative T lymphocytes of autoimmune MRL/lpr mice. PLoS One 2012; 7:e52161. [PMID: 23284917 PMCID: PMC3528777 DOI: 10.1371/journal.pone.0052161] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 11/12/2012] [Indexed: 12/12/2022] Open
Abstract
Lupus is a chronic inflammatory autoimmune disease influenced by multiple genetic loci including Fas Ligand (FasL) and P2X7 receptor (P2X7R). The Fas/Fas Ligand apoptotic pathway is critical for immune homeostasis and peripheral tolerance. Normal effector T lymphocytes up-regulate the transmembrane tyrosine phosphatase B220 before undergoing apoptosis. Fas-deficient MRL/lpr mice (lpr mutation) exhibit lupus and lymphoproliferative syndromes due to the massive accumulation of B220+ CD4–CD8– (DN) T lymphocytes. The precise ontogeny of B220+ DN T cells is unknown. B220+ DN T lymphocytes could be derived from effector CD4+ and CD8+ T lymphocytes, which have not undergone activation-induced cell death due to inactivation of Fas, or from a special cell lineage. P2X7R is an extracellular ATP-gated cell membrane receptor involved in the release of proinflammatory cytokines and TNFR1/Fas-independent cell death. P2X7R also regulate early signaling events involved in T-cell activation. We show herein that MRL/lpr mice carry a P2X7R allele, which confers a high sensitivity to ATP. However, during aging, the MRL/lpr T-cell population exhibits a drastically reduced sensitivity to ATP- or NAD-mediated stimulation of P2X7R, which parallels the increase in B220+ DN T-cell numbers in lymphoid organs. Importantly, we found that this B220+ DN T-cell subpopulation has a defect in P2X7R-mediated responses. The few B220+ T cells observed in normal MRL+/+ and C57BL/6 mice are also resistant to ATP or NAD treatment. Unexpectedly, while P2X7R mRNA and proteins are present inside of B220+ T cells, P2X7R are undetectable on the plasma membrane of these T cells. Our results prompt the conclusion that cell surface expression of B220 strongly correlates with the negative regulation of the P2X7R pathway in T cells.
Collapse
Affiliation(s)
- Sylvain M Le Gall
- Institut Jacques Monod, Centre National de la Recherche Scientifique, Université Paris Diderot, Paris, France
| | | | | | | | | | | | | |
Collapse
|
49
|
Takahashi M, Ota A, Karnan S, Hossain E, Konishi Y, Damdindorj L, Konishi H, Yokochi T, Nitta M, Hosokawa Y. Arsenic trioxide prevents nitric oxide production in lipopolysaccharide -stimulated RAW 264.7 by inhibiting a TRIF-dependent pathway. Cancer Sci 2012; 104:165-70. [PMID: 23106696 DOI: 10.1111/cas.12053] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 10/18/2012] [Accepted: 10/24/2012] [Indexed: 01/14/2023] Open
Abstract
Arsenic trioxide (ATO) is one of the most potent drugs in cancer chemotherapy, and is highly effective in treating both newly diagnosed and relapse patients with acute promyelocytic leukemia (APL). Despite a number of reports regarding the molecular mechanisms by which ATO promotes anti-tumor or pro-apoptotic activity in hematological and other solid malignancies, the effects of ATO on immune responses remain poorly understood. To further understand and clarify the effects of ATO on immune responses, we sought to examine whether ATO affects the production of nitric oxide (NO) in a lipopolysaccharide (LPS)-stimulated mouse macrophage cell line, RAW 264.7. Arsenic trioxide was found to prevent NO production in a dose-dependent manner. Arsenic trioxide significantly inhibited the increase in inducible nitric oxide synthase (iNOS) at both the mRNA and protein levels. Furthermore, our analyses revealed that the inhibitory effect of ATO on iNOS expression was ascribed to the prevention of IRF3 phosphorylation, interferon (IFN)-β expression, and STAT1 phosphorylation, but not the prevention of the MyD88-dependent pathway. Taken together, our results indicate that ATO prevents NO production by inhibiting the TIR-domain-containing adaptor protein inducing IFN-β (TRIF)-dependent pathway, thus highlighting an anti-inflammatory property of ATO in innate immunity.
Collapse
Affiliation(s)
- Miyuki Takahashi
- Department of Biochemistry, Department of Internal Medicine, Nagakute, Aichi, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Gesheva V, Szekeres Z, Mihaylova N, Dimitrova I, Nikolova M, Erdei A, Prechl J, Tchorbanov A. Generation of gene-engineered chimeric DNA molecules for specific therapy of autoimmune diseases. Hum Gene Ther Methods 2012; 23:357-65. [PMID: 23075110 PMCID: PMC4015069 DOI: 10.1089/hgtb.2012.051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 10/15/2012] [Indexed: 12/20/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the development of self-reactive B and T cells and autoantibody production. In particular, double-stranded DNA-specific B cells play an important role in lupus progression, and their selective elimination is a reasonable approach for effective therapy of SLE. DNA-based vaccines aim at the induction of immune response against the vector-encoded antigen. Here, we are exploring, as a new DNA-based therapy of SLE, a chimeric DNA molecule encoding a DNA-mimotope peptide, and the Fv but not the immunogenic Fc fragment of an FcγRIIb-specific monoclonal antibody. This DNA construct was inserted in the expression vector pNut and used as a naked DNA vaccine in a mouse model of lupus. The chimeric DNA molecule can be expressed in eukaryotic cells and cross-links cell surface receptors on DNA-specific B cells, delivering an inhibitory intracellular signal. Intramuscular administration of the recombinant DNA molecule to lupus-prone MRL/lpr mice prevented increase in IgG anti-DNA antibodies and was associated with a low degree of proteinuria, modulation of cytokine profile, and suppression of lupus nephritis.
Collapse
Affiliation(s)
- Vera Gesheva
- Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | | | | | | | | | | | | | | |
Collapse
|