1
|
Beene D, Miller C, Gonzales M, Kanda D, Francis I, Erdei E. Spatial nonstationarity and the role of environmental metal exposures on COVID-19 mortality in New Mexico. APPLIED GEOGRAPHY (SEVENOAKS, ENGLAND) 2024; 171:103400. [PMID: 39463888 PMCID: PMC11501077 DOI: 10.1016/j.apgeog.2024.103400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Worldwide, the COVID-19 pandemic has been influenced by a combination of environmental and sociodemographic drivers. To date, population studies have overwhelmingly focused on the impact of societal factors. In New Mexico, the rate of COVID-19 infection and mortality varied significantly among the state's geographically dispersed, and racially and ethnically diverse populations who are exposed to unique environmental contaminants related to resource extraction industries (e.g. fracking, mining, oil and gas exploration). By looking at local patterns of COVID-19 disease severity, we sought to uncover the spatially varying factors underlying the pandemic. We further explored the compounding role of potential long-term exposures to various environmental contaminants on COVID-19 mortality prior to widespread applications of vaccinations. To illustrate the spatial heterogeneity of these complex associations, we leveraged multiple modeling approaches to account for spatial non-stationarity in model terms. Multiscale geographically weighted regression (MGWR) results indicate that increased potential exposure to fugitive mine waste is significantly associated with COVID-19 mortality in areas of the state where socioeconomically disadvantaged populations were among the hardest hit in the early months of the pandemic. This relationship is paradoxically reversed in global models, which fail to account for spatial relationships between variables. This work contributes both to environmental health sciences and the growing body of literature exploring the implications of spatial nonstationarity in health research.
Collapse
Affiliation(s)
- Daniel Beene
- Community Environmental Health Program, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Geography & Environmental Studies, University of New Mexico, Albuquerque, NM, USA
| | - Curtis Miller
- Community Environmental Health Program, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Melissa Gonzales
- Department of Environmental Health Studies, Tulane University School of Public Health & Tropical Medicine, New Orleans, LA, USA
| | - Deborah Kanda
- Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Isaiah Francis
- Division of Epidemiology and Response, New Mexico Department of Health, Santa Fe, NM, USA
| | - Esther Erdei
- Community Environmental Health Program, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| |
Collapse
|
2
|
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
|
3
|
Omidifar N, Gholami A, Shokripour M, Nourani MA, Mohkam M, Mousavi SM, Hashemi SA, Khorram B, Ahmadabadi AN, Dara M. Protective Effects of Xanthine Derivatives Against Arsenic Trioxide-Induced Oxidative Stress in Mouse Hepatic and Renal Tissues. Drug Res (Stuttg) 2024; 74:133-144. [PMID: 38350632 DOI: 10.1055/a-2247-5232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
In this study, the protective efficacy of pentoxifylline (PTX) as a xanthine derivative against arsenic trioxide (ATO)-induced kidney and liver damage in mice was investigated. Thirty-six mice were divided into six groups, receiving intraperitoneal injections of saline, ATO, PTX, or a combination for four weeks. Blood samples were analyzed for serum biochemistry, while hepatic tissue underwent examination for histopathological changes and assessment of oxidative stress markers and antioxidant gene expression through Real-Time PCR. ATO exposure significantly increased serum markers (creatinine, ALT, BUN, ALP, AST) and induced histopathological changes in the liver. Moreover, it elevated renal and hepatic nitric oxide (NO) and lipid peroxidation (LPO) levels, and reduced antioxidant enzyme expression (CAT, GSR, GPx, MPO, SOD), total thiol groups (TTGs), and total antioxidant capacity (TAC). Conversely, PTX treatment effectively lowered serum hepatic and renal markers, improved antioxidant markers, and induced histopathological alterations. Notably, PTX did not significantly affect renal and hepatic NO levels. These findings suggest that PTX offers therapeutic potential in mitigating liver and acute kidney injuries induced by various insults, including exposure to ATO.
Collapse
Affiliation(s)
- Navid Omidifar
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Gholami
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mansoureh Shokripour
- Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Ali Nourani
- Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Mohkam
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyyed Mojtaba Mousavi
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei City, Taiwan
| | - Seyyed Alireza Hashemi
- Health Policy Research Center, Health Institute, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bagher Khorram
- Student Research Committee, School of Nursing and Midwifery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Nili Ahmadabadi
- Department of Pharmacology and Toxicology, Medicinal Plants and Natural Products Research Center, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan
| | - Mahintaj Dara
- Stem Cells and Transgenic Technology Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
4
|
Yin JZ, Shi XQ, Wang MD, Du H, Zhao XW, Li B, Yang MH. Arsenic trioxide elicits anti-tumor activity by inhibiting polarization of M2-like tumor-associated macrophages via Notch signaling pathway in lung adenocarcinoma. Int Immunopharmacol 2023; 117:109899. [PMID: 36827926 DOI: 10.1016/j.intimp.2023.109899] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/29/2023] [Accepted: 02/13/2023] [Indexed: 02/24/2023]
Abstract
Drug-resistant advanced lung adenocarcinoma (LUAD) is an aggressive malignancy with limited treatment options. A therapeutic strategy for drug-resistant LUAD is to target the tumor associated macrophages (TAMs), because they play an important role in tumor immune escape, progression and metastasis. In this study, we conducted in vivo and in vitro investigation of the inhibitory effect of arsenic trioxide (ATO) on polarization of TAMs educated by LUAD. We found that ATO at a concentration of 4 μM disrupted the Notch-dependent positive feedback loop between LUAD and TAMs. In this loop, ATO inhibited the expression of Jagged1 and Notch1 in LUAD and suppressed M2 polarization via down-regulating Notch-dependent paracrine of CCL2 and IL1β. As a result, the secretion of M2-derived TGF-β1 decreased, thus inducing inhibitions of LUAD proliferation, migration, invasion, colony formation and epithelial-mesenchymal transition. In xenograft mouse models, ATO significantly inhibited tumor growth and down-regulated infiltration of M2-like TAMs in tumor tissues. In clinical LUAD biopsy samples, high Jagged1/Notch1 expression positively correlated with tumor-infiltrated M2-like TAMs, leading to poor prognosis. In conclusion, our results identified a novel tumor immunomodulating function for ATO, which can inhibit the polarization of M2-type TAMs to exert anti-tumor effects in the tumor microenvironment. Our results demonstrated the translational potential of repurposing ATO to target TAMs for lung adenocarcinoma treatment.
Collapse
Affiliation(s)
- Ji-Zhong Yin
- Department of Respiratory and Critical Care Medicine, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai 200003, China
| | - Xiao-Qian Shi
- Department of Respiratory and Critical Care Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, 1279 Sanmen Road, Shanghai 200434, China
| | - Ming-Dong Wang
- Department of Thoracic Surgery, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, 1279 Sanmen Road, Shanghai 200434, China
| | - He Du
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, 507 Zhengmin Road, Shanghai 200433, China
| | - Xue-Wei Zhao
- Department of Thoracic Surgery, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, 1279 Sanmen Road, Shanghai 200434, China
| | - Bing Li
- Department of Respiratory and Critical Care Medicine, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai 200003, China; Department of Respiratory and Critical Care Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, 1279 Sanmen Road, Shanghai 200434, China.
| | - Meng-Hang Yang
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, 507 Zhengmin Road, Shanghai 200433, China.
| |
Collapse
|
5
|
Liu JL, Woo JMP, Parks CG, Costenbader KH, Jacobsen S, Bernatsky S. Systemic Lupus Erythematosus Risk: The Role of Environmental Factors. Rheum Dis Clin North Am 2022; 48:827-843. [PMID: 36332998 DOI: 10.1016/j.rdc.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Systemic lupus erythematosus (SLE) is a complex, chronic autoimmune disease. The etiology of SLE is multifactorial and includes potential environmental triggers, which may occur sequentially (the "multi-hit" hypothesis). This review focuses on SLE risk potentially associated with environmental factors including infections, the microbiome, diet, respirable exposures (eg, crystalline silica, smoking, air pollution), organic pollutants, heavy metals, and ultraviolet radiation.
Collapse
Affiliation(s)
- Jia Li Liu
- McGill University, Montreal, Quebec, Canada
| | - Jennifer M P Woo
- Epidemiology Branch, Department of Health and Human Services, National Institutes of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Christine G Parks
- Epidemiology Branch, Department of Health and Human Services, National Institutes of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Karen H Costenbader
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Søren Jacobsen
- Copenhagen Lupus and Vasculitis Clinic, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Sasha Bernatsky
- Centre for Outcomes Research and Evaluation, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| |
Collapse
|
6
|
Wang X, Xin B, Tan W, Xu Z, Li K, Li F, Zhong W, Peng S. DeepR2cov: deep representation learning on heterogeneous drug networks to discover anti-inflammatory agents for COVID-19. Brief Bioinform 2021; 22:6296505. [PMID: 34117734 PMCID: PMC8344611 DOI: 10.1093/bib/bbab226] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 05/14/2021] [Accepted: 05/24/2021] [Indexed: 02/06/2023] Open
Abstract
Recent studies have demonstrated that the excessive inflammatory response is an important factor of death in coronavirus disease 2019 (COVID-19) patients. In this study, we propose a deep representation on heterogeneous drug networks, termed DeepR2cov, to discover potential agents for treating the excessive inflammatory response in COVID-19 patients. This work explores the multi-hub characteristic of a heterogeneous drug network integrating eight unique networks. Inspired by the multi-hub characteristic, we design 3 billion special meta paths to train a deep representation model for learning low-dimensional vectors that integrate long-range structure dependency and complex semantic relation among network nodes. Based on the representation vectors and transcriptomics data, we predict 22 drugs that bind to tumor necrosis factor-α or interleukin-6, whose therapeutic associations with the inflammation storm in COVID-19 patients, and molecular binding model are further validated via data from PubMed publications, ongoing clinical trials and a docking program. In addition, the results on five biomedical applications suggest that DeepR2cov significantly outperforms five existing representation approaches. In summary, DeepR2cov is a powerful network representation approach and holds the potential to accelerate treatment of the inflammatory responses in COVID-19 patients. The source code and data can be downloaded from https://github.com/pengsl-lab/DeepR2cov.git.
Collapse
Affiliation(s)
- Xiaoqi Wang
- College of Computer Science and Electronic Engineering, Hunan University, China
| | - Bin Xin
- College of Computer Science and Electronic Engineering, Hunan University, China
| | - Weihong Tan
- Chinese Academy of Sciences in the College of Chemistry and Chemical Engineering, College of Biology, Hunan University, China
| | - Zhijian Xu
- Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China
| | - Kenli Li
- College of Computer Science and Electronic Engineering, Hunan University, China
| | - Fei Li
- Computer Network Information Center, Chinese Academy of Sciences, China
| | - Wu Zhong
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, China
| | - Shaoliang Peng
- College of Computer Science and Electronic Engineering, Hunan University, China
| |
Collapse
|
7
|
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: 2.8] [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
|
8
|
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: 17] [Impact Index Per Article: 3.4] [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
|
9
|
Erdei E, Shuey C, Pacheco B, Cajero M, Lewis J, Rubin RL. Elevated autoimmunity in residents living near abandoned uranium mine sites on the Navajo Nation. J Autoimmun 2019; 99:15-23. [PMID: 30878168 DOI: 10.1016/j.jaut.2019.01.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/15/2019] [Accepted: 01/17/2019] [Indexed: 10/27/2022]
Abstract
Specific autoantibodies were assessed among residents of the Navajo Nation in New Mexico chronically exposed to metal mixtures from uranium mine wastes and in drinking water supplies. Age and the extent of exposure to legacy waste from 100 abandoned uranium mine and mill sites were associated with antibodies to denatured DNA, previously known to be an early indicator of medication-induced autoimmunity. Surprisingly, autoantibodies to native DNA and/or chromatin were also linked to environmental exposure, specifically uranium consumption through drinking water for both men and women, while urinary arsenic was negatively associated with these autoantibodies in women. These findings suggest that contaminants derived from uranium mine waste enhanced development of autoantibodies in some individuals, while arsenic may be globally immunosuppressive with gender-specific effects. Specific autoantibodies may be a sensitive indicator of immune perturbation by environmental toxicants, an adverse effect not considered in current drinking water standards or regulatory risk assessment evaluations.
Collapse
Affiliation(s)
- Esther Erdei
- Community Environmental Health Program, Dept. of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, College of Pharmacy, 1 University of New Mexico, Albuquerque, NM 87131, USA
| | - Chris Shuey
- Southwest Research and Information Center, 105 Stanford Drive, SE, Albuquerque, NM 87106, USA
| | - Bernadette Pacheco
- Community Environmental Health Program, Dept. of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, College of Pharmacy, 1 University of New Mexico, Albuquerque, NM 87131, USA
| | - Miranda Cajero
- Community Environmental Health Program, Dept. of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, College of Pharmacy, 1 University of New Mexico, Albuquerque, NM 87131, USA
| | - Johnnye Lewis
- Community Environmental Health Program, Dept. of Pharmaceutical Sciences, University of New Mexico Health Sciences Center, College of Pharmacy, 1 University of New Mexico, Albuquerque, NM 87131, USA
| | - Robert L Rubin
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center School of Medicine, 1 University of New Mexico, Albuquerque, NM 87131, USA.
| |
Collapse
|
10
|
The Impact of Protein Acetylation/Deacetylation on Systemic Lupus Erythematosus. Int J Mol Sci 2018; 19:ijms19124007. [PMID: 30545086 PMCID: PMC6321219 DOI: 10.3390/ijms19124007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/30/2018] [Accepted: 12/10/2018] [Indexed: 02/08/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic inflammatory autoimmune disease in which the body’s immune system mistakenly attacks healthy cells. Although the exact cause of SLE has not been identified, it is clear that both genetics and environmental factors trigger the disease. Identical twins have a 24% chance of getting lupus disease if the other one is affected. Internal factors such as female gender and sex hormones, the major histocompatibility complex (MHC) locus and other genetic polymorphisms have been shown to affect SLE, as well as external, environmental influences such as sunlight exposure, smoking, vitamin D deficiency, and certain infections. Several studies have reported and proposed multiple associations between the alteration of the epigenome and the pathogenesis of autoimmune disease. Epigenetic factors contributing to SLE include microRNAs, DNA methylation status, and the acetylation/deacetylation of histone proteins. Additionally, the acetylation of non-histone proteins can also influence cellular function. A better understanding of non-genomic factors that regulate SLE will provide insight into the mechanisms that initiate and facilitate disease and also contribute to the development of novel therapeutics that can specifically target pathogenic molecular pathways.
Collapse
|
11
|
Yuan W, Chen J, Huang H, Cai Z, Ling Q, Huang F, Huang Z. Low-Dose Arsenic Trioxide Modulates the Differentiation of Mouse Embryonic Stem Cells. Chem Res Toxicol 2018; 31:472-481. [PMID: 29767511 DOI: 10.1021/acs.chemrestox.8b00027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Arsenic (As) is a well-known environmental pollutant, while arsenic trioxide (ATO) has been proven to be an effective treatment for acute promyelocytic leukemia, however, the mechanism underlying its dual effects is not fully understood. Embryonic stem cells (ESCs) exhibit properties of stemness and serve as a popular model to investigate epigenetic modifiers including environmental pollutants. Herein, the effects of low-dose ATO on differentiation were evaluated in vitro using a mouse ESCs (mESCs) cell line, CGR8. Cells treated with 0.2-0.5 μM ATO for 3-4 days had slight inhibition of proliferation with elevation of apoptosis, but obvious alterations of differentiation by morphological checking and alkaline phosphatase (AP) staining. Moreover, ATO exposure significantly decreased the mRNA expression of the stemness maintenance genes including Oct4, Nanog, and Rex-1 ( P < 0.01), whereas obviously increased some tissue-specific differentiation marker genes such as Gata4, Gata-6, AFP, and IHH. These alterations were consistent with the differentiation phenotype induced by retinoic acid (RA) and the expression patterns of distinct pluripotency markers such as SSEA-1 and Oct4. Furthermore, low-dose ATO led to a quantitative increase in Caspase 3 (CASP3) activation and subsequent cleavage of Nanog around 27 kDa, which corresponded with the mouse Nanog cleaved by CASP3 in a tube cleavage assay. Taken together, we suggest that low-dose ATO exposure will induce differentiation, other than apoptosis, of ESCs, such effects might be tuned partially by ATO-induced CASP3 activation and Nanog cleavage coupling with other differentiation related genes involved. The present findings provide a preliminary action mechanism of arsenic on the cell fate determination.
Collapse
Affiliation(s)
- Wenlin Yuan
- Department of Biotechnology, School of Life Science and Technology , Jinan University , Guangzhou 510632 , Guangdong Province , China
| | - Jun Chen
- Department of Biotechnology, School of Life Science and Technology , Jinan University , Guangzhou 510632 , Guangdong Province , China
| | - Hongren Huang
- Department of Biotechnology, School of Life Science and Technology , Jinan University , Guangzhou 510632 , Guangdong Province , China
| | - Zhihui Cai
- Department of Biotechnology, School of Life Science and Technology , Jinan University , Guangzhou 510632 , Guangdong Province , China
| | - Qinjie Ling
- Department of Biotechnology, School of Life Science and Technology , Jinan University , Guangzhou 510632 , Guangdong Province , China
| | - Feng Huang
- Department of Rehabilitation Medicine, School of Medical Engineering , Foshan University , Foshan 528000 , Guangdong Province , China
| | - Zhi Huang
- Department of Biotechnology, School of Life Science and Technology , Jinan University , Guangzhou 510632 , Guangdong Province , China
| |
Collapse
|