1
|
Zhang W, Wang Z, Fu Y, Thakur C, Ji H, Bi Z, Qiu Y, Elangbam M, Haley J, Chen F. Aryl Hydrocarbon Receptor (AHR) Suppresses Arsenic (As 3+)-Induced Malignant Transformation by Antagonizing TOX Expression. Int J Biol Sci 2025; 21:2747-2761. [PMID: 40303305 PMCID: PMC12035900 DOI: 10.7150/ijbs.107268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/17/2025] [Indexed: 05/02/2025] Open
Abstract
Environmental arsenic (As³⁺) exposure poses a significant public health concern due to its carcinogenic potential. Our previous research suggests that As³⁺-induced carcinogenesis is mediated by inhibition of the aryl hydrocarbon receptor (AHR). However, the precise role of AHR in As³⁺-induced malignant transformation as well as cancer stem-like cell (CSC) formation, along with its underlying mechanisms, remains unclear. In this study, we used BEAS-2B cells with CRISPR-Cas9 gene editing, RNA sequencing, and immunoprecipitation to examine AHR's role in As³⁺-induced CSC development. Our findings reveal that AHR suppresses CSC formation triggered by low-dose As³⁺ (0.5 μM) via transcriptional repression of TOX, a high mobility group box DNA binding protein that play a critical role in T cell exhaustion within tumor immunology. TOX knockdown inhibited CSC formation, while its overexpression enhanced cMYC, a CSC-associated transcription factor. TOX interactome analysis identified associations with proteins such as KCTD10, TRIM21, HMGA1, FLOT1, and FLOT2, which may regulate TOX's stability and activity. Enrichment analyses highlighted their involvement in cancer-related pathways, supporting the role of TOX in promoting CSC formation during As³⁺-induced carcinogenesis. Notably, this study identifies TOX as an oncogenic factor in non-immunological contexts and underscores AHR's tumor-suppressive function through TOX repression, offering novel insights into the mechanisms underlying As³⁺-induced carcinogenesis.
Collapse
Affiliation(s)
| | - Ziwei Wang
- Stony Brook Cancer Center, Department of Pathology, Renaissance School of Medicine, Stony Brook University. Lauterbur Drive, Stony Brook, NY 11794, USA
| | | | | | | | | | | | | | | | - Fei Chen
- Stony Brook Cancer Center, Department of Pathology, Renaissance School of Medicine, Stony Brook University. Lauterbur Drive, Stony Brook, NY 11794, USA
| |
Collapse
|
2
|
Wu X, Kreutz A, Dixon D, Tokar EJ. Engineering human cerebral organoids to explore mechanisms of arsenic-induced developmental neurotoxicity. Toxicol Appl Pharmacol 2025; 496:117230. [PMID: 39842615 PMCID: PMC11846691 DOI: 10.1016/j.taap.2025.117230] [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: 10/16/2024] [Revised: 12/26/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025]
Abstract
Modeling brain development and function is challenging due to complexity of the organ. Human pluripotent stem cell (PSC)-derived brain-like organoids provide new tools to study the human brain. Compared with traditional in vivo toxicological studies, these 3D models, together with 2D cellular assays, enhance our understanding of the mechanisms of developmental neurotoxicity (DNT) during the early stages of neurogenesis and offer numerous advantages including a rapid, cost-effective approach for understanding compound mechanisms and assessing chemical safety. Arsenic (As) exposure is associated with DNT, although the mechanisms involved are not well-defined. Here, we used 3D PSC-derived embryoid bodies (EBs) to recapitulate events involved in embryogenesis and neurogenesis before neural induction, and EB-derived cerebral organoids to mimic neural development in vivo. As (0.5 μM; 35 ppb) increased ectoderm differentiation within the EBs by upregulating genes (PAX6, SOX1) critical for embryonic development. Histological staining of EBs showed As disrupted neural rosette structures. qPCR and RNA-seq showed As inhibited expression of markers of mature neural cells (MAP2+/vGLUT2+) and astrocytes (GFAP+). In organoids, Ingenuity Pathway Analysis was used to identify the top 5 pathways affected by As exposure, and Gene Ontology enrichment analysis found several key signaling pathways to be inhibited by As exposure. These data provide insights into pathways contributing to As-induced inhibition of neurite outgrowth and disrupted neural rosette structures in the 2D neurite outgrowth assay and in organoids, respectively. Results herein show this multipronged 2D/3D approach can provide valuable insights into cellular events and molecular mechanisms of As-induced DNT.
Collapse
Affiliation(s)
- Xian Wu
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, United States of America
| | - Anna Kreutz
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, United States of America
| | - Darlene Dixon
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, United States of America
| | - Erik J Tokar
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, United States of America.
| |
Collapse
|
3
|
Garg A, Bandyopadhyay S. Role of an interdependent Wnt, GSK3-β/β-catenin and HB-EGF/EGFR mechanism in arsenic-induced hippocampal neurotoxicity in adult mice. CHEMOSPHERE 2024; 352:141375. [PMID: 38325618 DOI: 10.1016/j.chemosphere.2024.141375] [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: 09/04/2023] [Revised: 01/17/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
We previously reported the neurotoxic effects of arsenic in the hippocampus. Here, we explored the involvement of Wnt pathway, which contributes to neuronal functions. Administering environmentally relevant arsenic concentrations to postnatal day-60 (PND60) mice demonstrated a dose-dependent increase in hippocampal Wnt3a and its components, Frizzled, phospho-LRP6, Dishevelled and Axin1 at PND90 and PND120. However, p-GSK3-β(Ser9) and β-catenin levels although elevated at PND90, decreased at PND120. Additionally, treatment with Wnt-inhibitor, rDkk1, reduced p-GSK3-β(Ser9) and β-catenin at PND90, but failed to affect their levels at PND120, indicating a time-dependent link with Wnt. To explore other underlying factors, we assessed epidermal growth factor receptor (EGFR) pathway, which interacts with GSK3-β and appears relevant to neuronal functions. We primarily found that arsenic reduced hippocampal phosphorylated-EGFR and its ligand, Heparin-binding EGF-like growth factor (HB-EGF), at both PND90 and PND120. Moreover, treatment with HB-EGF rescued p-GSK3-β(Ser9) and β-catenin levels at PND120, suggesting their HB-EGF/EGFR-dependent regulation at this time point. Additionally, rDkk1, LiCl (GSK3-β-activity inhibitor), or β-catenin protein treatments induced a time-dependent recovery in HB-EGF, indicating potential inter-dependent mechanism between hippocampal Wnt/β-catenin and HB-EGF/EGFR following arsenic exposure. Fluorescence immunolabeling then validated these findings in hippocampal neurons. Further exploration of hippocampal neuronal survival and apoptosis demonstrated that treatment with rDkk1, LiCl, β-catenin and HB-EGF improved Nissl staining and NeuN levels, and reduced cleaved-caspase-3 levels in arsenic-treated mice. Supportively, we detected improved Y-Maze and Passive Avoidance performances for learning-memory functions in these mice. Overall, our study provides novel insights into Wnt/β-catenin and HB-EGF/EGFR pathway interaction in arsenic-induced hippocampal neurotoxicity.
Collapse
Affiliation(s)
- Asmita Garg
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sanghamitra Bandyopadhyay
- Systems Toxicology Group, Food, Drug & Chemical, Environment and Systems Toxicology Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
4
|
Park HR, Azzara D, Cohen ED, Boomhower SR, Diwadkar AR, Himes BE, O'Reilly MA, Lu Q. Identification of novel NRF2-dependent genes as regulators of lead and arsenic toxicity in neural progenitor cells. JOURNAL OF HAZARDOUS MATERIALS 2024; 463:132906. [PMID: 37939567 PMCID: PMC10842917 DOI: 10.1016/j.jhazmat.2023.132906] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023]
Abstract
Lead (Pb) and arsenic (As) are prevalent metal contaminants in the environment. Exposures to these metals are associated with impaired neuronal functions and adverse effects on neurodevelopment in children. However, the molecular mechanisms by which Pb and As impair neuronal functions remain poorly understood. Here, we identified F2RL2, TRIM16L, and PANX2 as novel targets of Nuclear factor erythroid 2-related factor 2 (NRF2)-the master transcriptional factor for the oxidative stress response-that are commonly upregulated with both Pb and As in human neural progenitor cells (NPCs). Using a ChIP (Chromatin immunoprecipitation)-qPCR assay, we showed that NRF2 directly binds to the promoter region of F2RL2, TRIM16L, and PANX2 to regulate expression of these genes. We demonstrated that F2RL2, PANX2, and TRIM16L have differential effects on cell death, proliferation, and differentiation of NPCs in both the presence and absence of metal exposures, highlighting their roles in regulating NPC function. Furthermore, the analyses of the transcriptomic data on NPCs derived from autism spectrum disorder (ASD) patients revealed that dysregulation of F2RL2, TRIM16L, and PANX2 was associated with ASD genetic backgrounds and ASD risk genes. Our findings revealed that Pb and As induce a shared NRF2-dependent transcriptional response in NPCs and identified novel genes regulating NPC function. While further in vivo studies are warranted, this study provides a novel mechanism linking metal exposures to NPC function and identifies potential genes of interest in the context of neurodevelopment.
Collapse
Affiliation(s)
- Hae-Ryung Park
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA.
| | - David Azzara
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Ethan D Cohen
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Steven R Boomhower
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Avantika R Diwadkar
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Blanca E Himes
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael A O'Reilly
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Quan Lu
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
5
|
Clemens Z, Wang K, Ambrosio F, Barchowsky A. Arsenic disrupts extracellular vesicle-mediated signaling in regenerating myofibers. Toxicol Sci 2023; 195:231-245. [PMID: 37527016 PMCID: PMC10535782 DOI: 10.1093/toxsci/kfad075] [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] [Indexed: 08/03/2023] Open
Abstract
Chronic exposure to environmental arsenic is a public health crisis affecting hundreds of millions of individuals worldwide. Though arsenic is known to contribute to many pathologies and diseases, including cancers, cardiovascular and pulmonary diseases, and neurological impairment, the mechanisms for arsenic-promoted disease remain unresolved. This is especially true for arsenic impacts on skeletal muscle function and metabolism, despite the crucial role that skeletal muscle health plays in maintaining cardiovascular health, systemic homeostasis, and cognition. A barrier to researching this area is the challenge of interrogating muscle cell-specific effects in biologically relevant models. Ex vivo studies investigating mechanisms for muscle-specific responses to arsenic or other environmental contaminants primarily utilize traditional 2-dimensional culture models that cannot elucidate effects on muscle physiology or function. Therefore, we developed a contractile 3-dimensional muscle construct model-composed of primary mouse muscle progenitor cells differentiated in a hydrogel matrix-to study arsenic exposure impacts on skeletal muscle regeneration. Muscle constructs exposed to low-dose (50 nM) arsenic exhibited reduced strength and myofiber diameter following recovery from muscle injury. These effects were attributable to dysfunctional paracrine signaling mediated by extracellular vesicles (EVs) released from muscle cells. Specifically, we found that EVs collected from arsenic-exposed muscle constructs recapitulated the inhibitory effects of direct arsenic exposure on myofiber regeneration. In addition, muscle constructs treated with EVs isolated from muscles of arsenic-exposed mice displayed significantly decreased strength. Our findings highlight a novel model for muscle toxicity research and uncover a mechanism of arsenic-induced muscle dysfunction by the disruption of EV-mediated intercellular communication.
Collapse
Affiliation(s)
- Zachary Clemens
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Kai Wang
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, Massachusetts, USA
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA
| | - Fabrisia Ambrosio
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, Massachusetts, USA
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Boston, Massachusetts, USA
| | - Aaron Barchowsky
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
6
|
Haidar Z, Fatema K, Shoily SS, Sajib AA. Disease-associated metabolic pathways affected by heavy metals and metalloid. Toxicol Rep 2023; 10:554-570. [PMID: 37396849 PMCID: PMC10313886 DOI: 10.1016/j.toxrep.2023.04.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/21/2023] [Accepted: 04/23/2023] [Indexed: 07/04/2023] Open
Abstract
Increased exposure to environmental heavy metals and metalloids and their associated toxicities has become a major threat to human health. Hence, the association of these metals and metalloids with chronic, age-related metabolic disorders has gained much interest. The underlying molecular mechanisms that mediate these effects are often complex and incompletely understood. In this review, we summarize the currently known disease-associated metabolic and signaling pathways that are altered following different heavy metals and metalloids exposure, alongside a brief summary of the mechanisms of their impacts. The main focus of this study is to explore how these affected pathways are associated with chronic multifactorial diseases including diabetes, cardiovascular diseases, cancer, neurodegeneration, inflammation, and allergic responses upon exposure to arsenic (As), cadmium (Cd), chromium (Cr), iron (Fe), mercury (Hg), nickel (Ni), and vanadium (V). Although there is considerable overlap among the different heavy metals and metalloids-affected cellular pathways, these affect distinct metabolic pathways as well. The common pathways may be explored further to find common targets for treatment of the associated pathologic conditions.
Collapse
|
7
|
Perego MC, McMichael BD, Bain LJ. Arsenic impairs stem cell differentiation via the Hippo signaling pathway. Toxicol Res (Camb) 2023; 12:296-309. [PMID: 37125325 PMCID: PMC10141767 DOI: 10.1093/toxres/tfad018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 04/03/2023] Open
Abstract
Arsenic is a ubiquitous toxic metalloid, with over 150 million people exposed to arsenic concentrations above the current 10 ppb drinking water standard through contaminated food and water. Arsenic is a known developmental toxicant as neuronal and muscle development are disrupted following arsenic exposure during embryogenesis. In this study, murine embryonic stem cells were chronically exposed to 0.1 μM (7.5 ppb) arsenic for 32 weeks. RNA sequencing showed that the Hippo signaling pathway, which is involved in embryonic development and pluripotency maintenance, is impaired following arsenic exposure. Thus, temporal changes in the Hippo pathway's core components and its downstream target genes Ctgf and c-Myc were investigated. Protein expression of the pathway's main effector YAP in its active form was significantly upregulated by 3.7-fold in arsenic-exposed cells at week 8, while protein expression of inactive phosphorylated YAP was significantly downregulated by 2.5- and 2-fold at weeks 8 and 16. Exposure to arsenic significantly increased the ratio between nuclear and cytoplasmic YAP by 1.9-fold at weeks 16 and 28. The ratio between nuclear and cytoplasmic transcriptional enhancer factor domain was similarly increased in arsenic-treated samples by 3.4- and 1.6-fold at weeks 16 and 28, respectively. Levels of Ctgf and c-Myc were also upregulated following arsenic exposure. These results suggest that chronic exposure to an environmentally relevant arsenic concentration might hinder cellular differentiation and maintain pluripotency through the impairment of the Hippo signaling pathway resulting in increased YAP activation.
Collapse
Affiliation(s)
- M Chiara Perego
- Department of Biological Sciences, Clemson University, 132 Long Hall, Clemson, SC, 29631, United States
| | - Benjamin D McMichael
- Department of Biological Sciences, Clemson University, 132 Long Hall, Clemson, SC, 29631, United States
- Department of Biology, University of North Carolina, 120 South Road, Chapel Hill, NC, 27599, United States
| | - Lisa J Bain
- Department of Biological Sciences, Clemson University, 132 Long Hall, Clemson, SC, 29631, United States
| |
Collapse
|
8
|
Kellett MP, Jatko JT, Darling CL, Ventrello SW, Bain LJ. Arsenic Exposure Impairs Intestinal Stromal Cells. Toxicol Lett 2022; 361:54-63. [PMID: 35378173 PMCID: PMC9038714 DOI: 10.1016/j.toxlet.2022.03.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/23/2022] [Accepted: 03/17/2022] [Indexed: 01/01/2023]
Abstract
Arsenic is a toxicant commonly found in drinking water. Even though its main route of exposure is oral, little is known of the impact of in vivo arsenic exposure on small intestine. In vitro studies have shown that arsenic decreases differentiation of stem and progenitor cells in several different tissues. Thus, small intestinal organoids were used to assess if arsenic exposure would also impair intestinal stem cell differentiation. Unexpectedly, no changes in markers of differentiated epithelial cells were seen. However, exposing mice to 100 ppb arsenic in drinking water for 5 weeks impaired distinct populations of intestinal stromal cells. Arsenic reduced the width of the pericryptal lamina propria by 1.6-fold, and reduced Pdgfra mRNA expression, which is expressed in intestinal telocytes and trophocytes, by 4.2-fold. The height or extension of Pdgfra+ telopodes into the villus tip was also significantly reduced. Transcript expression of several other stromal cell markers, such as Grem1, Gli, CD81, were reduced by 1.9-, 2.3-, and 1.4-fold, respectively. Further, significant correlations exist between levels of Pdgfra and Gli1, Grem1, and Bmp4. Our results suggest arsenic impairs intestinal trophocytes and telocytes, leading to alterations in the Bmp signaling pathway.
Collapse
|
9
|
Flanigan TJ, Ferguson SA, Law CD, Rosas-Hernandez H, Cuevas-Martinez E, Fitzpatrick S, Shen AN. Neurobehavioral and neurochemical effects of perinatal arsenite exposure in Sprague-Dawley rats. Neurotoxicol Teratol 2021; 90:107059. [PMID: 34979254 DOI: 10.1016/j.ntt.2021.107059] [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: 08/17/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 11/19/2022]
Abstract
Exposure to relatively high levels of inorganic arsenic (iAs) is associated with detrimental effects on human health, including cancer and diabetes. The effects of lower-level exposures are less clear, and gaps in the literature exist as to the effects of iAs exposure on neurodevelopment. The current study assessed the effects of perinatal iAs exposure on rodent neurodevelopment and behavior. Pregnant Sprague-Dawley (SD) rats were exposed to arsenite (AsIII) via oral gavage on gestational days (GD) 6 through 21, and pups were directly dosed via gavage on postnatal days (PND) 1 through 21. Dams and offspring received the same doses: 0.00, 0.10, 1.50, or 3.75 mg/kg/day. Male and female offspring underwent a battery of behavioral assessments from weaning until PND 180. Brain arsenic levels increased in a dose-dependent manner at both PND 1 and 21. Results from the behavioral tests show that pre- and postnatal AsIII exposure did not adversely affect offspring weight gain, adolescent motor and cognitive functions, or adult motor and cognitive functions in the SD rat. There were no differences in concentration of several brain proteins associated with blood-brain barrier permeability, dopamine functions, and inflammation.
Collapse
Affiliation(s)
- Timothy J Flanigan
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR 72079, United States of America
| | - Sherry A Ferguson
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR 72079, United States of America
| | - Charles D Law
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR 72079, United States of America
| | - Hector Rosas-Hernandez
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR 72079, United States of America
| | - Elvis Cuevas-Martinez
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR 72079, United States of America
| | - Suzanne Fitzpatrick
- Office of the Center Director, Center for Food Safety and Applied Nutrition/FDA, , United States of America
| | - Andrew N Shen
- Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR 72079, United States of America.
| |
Collapse
|
10
|
Jatko JT, Darling CL, Kellett MP, Bain LJ. Arsenic exposure in drinking water reduces Lgr5 and secretory cell marker gene expression in mouse intestines. Toxicol Appl Pharmacol 2021; 422:115561. [PMID: 33957193 PMCID: PMC11931411 DOI: 10.1016/j.taap.2021.115561] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 02/28/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022]
Abstract
Arsenic is a global health concern that causes toxicity through ingestion of contaminated water and food. In vitro studies suggest that arsenic reduces stem and progenitor cell differentiation. Thus, this study determined if arsenic disrupted intestinal stem cell (ISC) differentiation, thereby altering the number, location, and/or function of intestinal epithelial cells. Adult male C57BL/6 mice were exposed to 0 or 100 ppb sodium arsenite (AsIII) through drinking water for 5 weeks. Duodenal sections were collected to assess changes in morphology, proliferation, and cell types. qPCR analysis revealed a 40% reduction in Lgr5 transcripts, an ISC marker, in the arsenic-exposed mice, although there were no changes in the protein expression of Olfm4. Secretory cell-specific transcript markers of Paneth (Defa1), Goblet (Tff3), and secretory transit amplifying (Math1) cells were reduced by 51%, 44%, and 30% respectively, in the arsenic-exposed mice, indicating significant impacts on the Wnt-dependent differentiation pathway. Further, protein levels of phosphorylated β-catenin were reduced in the arsenic-exposed mice, which increased the expression of Wnt-dependent transcripts CD44 and c-myc. PCA analysis, followed by MANOVA and regression analyses, revealed significant changes and correlations between Lgr5 and the transit amplifying (TA) cell markers Math1 and Hes1, which are in the secretory cell pathway. Similar comparisons between Math1 and Defa1 show that terminal differentiation into Paneth cells is also reduced in the arsenic-exposed mice. The data suggests that ISCs are not lost following arsenic exposure, but rather, specific Wnt-dependent progenitor cell formation and terminal differentiation in the small intestine is reduced.
Collapse
Affiliation(s)
- Jordan T Jatko
- Environmental Toxicology Graduate Program, Clemson University, 132 Long Hall, Clemson, SC 29634, USA
| | - Caitlin L Darling
- Department of Biological Sciences, Clemson University, 132 Long Hall, Clemson, SC 29634, USA
| | - Michael P Kellett
- Department of Biological Sciences, Clemson University, 132 Long Hall, Clemson, SC 29634, USA
| | - Lisa J Bain
- Environmental Toxicology Graduate Program, Clemson University, 132 Long Hall, Clemson, SC 29634, USA; Department of Biological Sciences, Clemson University, 132 Long Hall, Clemson, SC 29634, USA.
| |
Collapse
|
11
|
Xie L, Hu WY, Hu DP, Shi G, Li Y, Yang J, Prins GS. Effects of Inorganic Arsenic on Human Prostate Stem-Progenitor Cell Transformation, Autophagic Flux Blockade, and NRF2 Pathway Activation. ENVIRONMENTAL HEALTH PERSPECTIVES 2020; 128:67008. [PMID: 32525701 PMCID: PMC7289393 DOI: 10.1289/ehp6471] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/18/2020] [Accepted: 05/06/2020] [Indexed: 05/26/2023]
Abstract
BACKGROUND Inorganic arsenic (iAs) is an environmental toxicant associated with an increased risk of prostate cancer in chronically exposed populations worldwide. However, the biological mechanisms underlying iAs-induced prostate carcinogenesis remain unclear. OBJECTIVES We studied how iAs affects normal human prostate stem-progenitor cells (PrSPCs) and drives transformation and interrogated the molecular mechanisms involved. METHODS PrSPCs were enriched by spheroid culture from normal human primary or immortalized prostate epithelial cells, and their differentiation capability was evaluated by organoid culture. Microarray analysis was conducted to identify iAs-dysregulated genes, and lentiviral infection was used for stable manipulation of identified genes. Soft agar colony growth assays were applied to examine iAs-induced transformation. For in vivo study, PrSPCs mixed with rat urogenital sinus mesenchyme were grafted under the renal capsule of nude mice to generate prostatelike tissues, and mice were exposed to 5 ppm (∼65μM) iAs in drinking water for 3 months. RESULTS Low-dose iAs (1μM) disturbed PrSPC homeostasis in vitro, leading to increased self-renewal and suppressed differentiation. Transcriptomic analysis indicated that iAs activated oncogenic pathways in PrSPCs, including the KEAP1-NRF2 pathway. Further, iAs-exposed proliferative progenitor cells exhibited NRF2 pathway activation that was sustained in their progeny cells. Knockdown of NRF2 inhibited spheroid formation by driving PrSPC differentiation, whereas its activation enhanced spheroid growth. Importantly, iAs-induced transformation was suppressed by NRF2 knockdown. Mechanistically, iAs suppressed Vacuolar ATPase subunit VMA5 expression, impairing lysosome acidification and inhibiting autophagic protein degradation including p62, which further activated NRF2. In vivo, chronic iAs exposure activated NRF2 in both epithelial and stroma cells of chimeric human prostate grafts and induced premalignant events. CONCLUSIONS Low-dose iAs increased self-renewal and decreased differentiation of human PrSPCs by activating the p62-NRF2 axis, resulting in epithelial cell transformation. NRF2 is activated by iAs through specific autophagic flux blockade in progenitor cells, which may have potential therapeutic implications. https://doi.org/10.1289/EHP6471.
Collapse
Affiliation(s)
- Lishi Xie
- Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
- Chicago Center for Health and Environment, Chicago, Illinois, USA
| | - Wen-Yang Hu
- Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
- Chicago Center for Health and Environment, Chicago, Illinois, USA
| | - Dan-Ping Hu
- Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Guangbin Shi
- Division of Cardiothoracic Surgery, The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Ye Li
- Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Jianfu Yang
- Department of Urology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
| | - Gail S. Prins
- Department of Urology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
- Chicago Center for Health and Environment, Chicago, Illinois, USA
- Departments of Physiology & Biophysics and Pathology, College of Medicine; Division of Epidemiology & Biostatistics, School of Public Health, University of Illinois at Chicago, Chicago, Illinois, USA
- University of Illinois Cancer Center, Chicago, Illinois, USA
| |
Collapse
|
12
|
Li S, Ren Q. Effects of Arsenic on wnt/β-catenin Signaling Pathway: A Systematic Review and Meta-analysis. Chem Res Toxicol 2020; 33:1458-1467. [PMID: 32307979 DOI: 10.1021/acs.chemrestox.0c00019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
We aimed to systematically evaluate the regulatory effect of arsenic on wnt/β-catenin signaling pathway and to provide theoretical basis for revealing the mechanism of the relationship between arsenic and cell proliferation. The meta-analysis was carried out using Revman5.2 and Stata13.0 to describe the differences between groups with standard mean difference. We found in normal cells that the levels of wnt3a, β-catenin, glycogen synthase kinase-3β phosphorylated at serine 9 (p-GSK-3β(Ser9)), cyclinD1, proto-oncogene c-myc, and vascular endothelial growth factor (VEGF) in the arsenic intervention group were higher than those in the control group, and the level of glycogen synthase kinase-3β (GSK-3β) was lower than that in the control group (P < 0.05, respectively). Subgroup analysis showed that for a long time period (>24 h), the level of β-catenin in the arsenic intervention group was higher than that in the control group, and the level of GSK-3β of the same long-time period (>24 h) with low-dose (≤5 μM) intervention was lower than those in the control group (P < 0.05, respectively). In cancer cells, the levels of β-catenin, cyclinD1, c-myc, and VEGF in the arsenic intervention group were lower than those in the control group, while the level of GSK-3β in the arsenic intervention group was higher than that in the control group (P < 0.05, respectively). Subgroup analysis showed that the levels of β-catenin, cyclinD1, and c-myc in the high-dose (>5 μM) arsenic intervention group were lower than those in the control group, and the levels of β-catenin and cyclinD1 in the high-dose (>5 μM) arsenic intervention group were lower than those in the low-dose (≤5 μM) arsenic intervention group (P < 0.05, respectively). In addition, the regulation of arsenic on β-catenin was dose-dependent in the range of arsenic concentration from 0 to 7.5 μM. This study revealed that arsenic could upregulate wnt/β-catenin signaling pathway in normal cells and downregulate it in cancer cells, and its effect was affected by time and dose.
Collapse
Affiliation(s)
- Shugang Li
- Department of Child, Adolescent Health and Maternal Health, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Qingxin Ren
- Department of Public Health, College of Medicine, Shihezi University, Shihezi 832000, Xinjiang China
| |
Collapse
|
13
|
Bi Z, Zhang Q, Fu Y, Wadgaonkar P, Zhang W, Almutairy B, Xu L, Rice M, Qiu Y, Thakur C, Chen F. Nrf2 and HIF1α converge to arsenic-induced metabolic reprogramming and the formation of the cancer stem-like cells. Theranostics 2020; 10:4134-4149. [PMID: 32226544 PMCID: PMC7086359 DOI: 10.7150/thno.42903] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 02/06/2020] [Indexed: 01/02/2023] Open
Abstract
In this report, we demonstrated that inorganic arsenic (iAs) induces generation of the cancer stem-like cells (CSCs) through Nrf2-dependent HIF1α activation, and the subsequent metabolic reprogramming from mitochondrial oxidative phosphorylation to glycolysis in epithelial cells. Methods: Genome-wide ChIP-seq analysis was performed to investigate the global binding of Nrf2 and/or HIF1α on the genome in the cells treated with iAs. Both untargeted metabolomics and UDP-13C-glucose flux were applied to determine metabolic reprogramming in the iAs-induced CSCs. The role of Nrf2 on iAs-induced HIF1α and other stemness gene expression was validated by lentiviral transfection of Nrf2 inhibitor Keap1 and CRISPR-Cas9-mediated Nrf2 gene knockout, respectively. Results: The CSCs induced by iAs exhibit a diminished mitochondrial oxidative phosphorylation and an enhanced glycolysis that is actively shunted to the hexosamine biosynthetic pathway (HBP) and serine/glycine pathway. ChIP-seq data revealed that treatment of the cells with iAs amplified Nrf2 enrichment peaks in intergenic region, promoter and gene body. In contrast, a shift of the HIF1α peaks from distal intergenic region to gene promoter and the first exon was noted. Both Nrf2 and HIF1α are responsible for the iAs-induced expression of the glycolytic genes and the genes important for the stemness of the CSCs. Intriguingly, we also discovered a mutual transcriptional regulation between Nrf2 and HIF1α. Inhibition of Nrf2 by lentiviral infection of Keap1, or knockout of Nrf2 by CRISPR-Cas9 gene editing, not only blocked iAs-induced HIF1α activation, but reduced the expression of the key stemness genes for the formation of CSCs also. Conclusion: We demonstrated that Nrf2 activation is an initiating signal for iAs-induced HIF1α activation, and Nrf2 and HIF1α played a concerted role on inducing metabolic reprogramming and the CSCs.
Collapse
Affiliation(s)
- Zhuoyue Bi
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
- School of Health Sciences, Wuhan University, 115 Donghu Road, Wuhan 430071, China
- Hubei Provincial Key Laboratory of Applied Toxicology, Hubei Provincial Center for Disease Control and Prevention, 8 Zhudaoquanbei Road, Wuhan 430079, China
| | - Qian Zhang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| | - Yao Fu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| | - Priya Wadgaonkar
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| | - Wenxuan Zhang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| | - Bandar Almutairy
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
- College of Pharmacy, Al-Dawadmi Campus, Shaqra University, P.O.Box 11961, Riyadh, Kingdom of Saudi Arabia
| | - Liping Xu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| | - M'Kya Rice
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| | - Yiran Qiu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| | - Chitra Thakur
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| | - Fei Chen
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201, USA
| |
Collapse
|
14
|
Arsenic hampered embryonic development: An in vivo study using local Bangladeshi Danio rerio model. Toxicol Rep 2020; 7:155-161. [PMID: 31993334 PMCID: PMC6976906 DOI: 10.1016/j.toxrep.2019.12.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 10/23/2019] [Accepted: 12/29/2019] [Indexed: 11/20/2022] Open
Abstract
Exposure to arsenic results delayed and deformed embryonic development. Arsenic exposure increased the mortality rate of embryos. Arsenic exposure may increase miscarriage or abortion rate in the pregnant mother.
Zebrafish (Danio rerio) has appeared as a valuable and popular model species to study the developmental and toxicological impact of environmental pollutants. To get insights on the toxicological effect of arsenic on early embryonic development, a controlled breeding of local Bangladeshi zebrafish followed by comprehensive microscopic analysis was conducted to study the embryonic development after exposure to different concentrations of arsenic ranges from 4−120 h post-fertilization. Zebrafish embryos exposed to 2 mM of arsenic displayed distinguishable developmental delay compared to control. At three days post-fertilization, a distinct phenotype appears in arsenic-treated embryos, which can be characterized by dechorionated embryos, larger egg mass, pericardial edema, abnormal heart rate, and abnormal head development. Remarkably, the death rate of the arsenic-treated embryos was significantly higher compared to control. Collectively, these findings indicate that exposure to arsenic may result in abnormal embryonic development. These results suggest for proper management of the pregnant mother in the arsenic-exposed area, and may also explain the incidence of increased miscarriage/abortion rate in arsenic water drinking pregnant mother.
Collapse
|
15
|
Worley JR, Parker GC. Effects of environmental stressors on stem cells. World J Stem Cells 2019; 11:565-577. [PMID: 31616535 PMCID: PMC6789190 DOI: 10.4252/wjsc.v11.i9.565] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 07/12/2019] [Accepted: 09/04/2019] [Indexed: 02/06/2023] Open
Abstract
Environmental toxicants are ubiquitous, and many are known to cause harmful health effects. However, much of what we know or think we know concerning the targets and long-term effects of exposure to environmental stressors is sadly lacking. Toxicant exposure may have health effects that are currently mischaracterized or at least mechanistically incompletely understood. While much of the recent excitement about stem cells (SCs) focuses on their potential as therapeutic agents, they also offer a valuable resource to give us insight into the mechanisms and risks of toxicant effects. Not only as a response to the increasing ethical pressure to reduce animal testing, SC studies allow us valuable insight into the true effects of human exposure to environmental stressors under controlled conditions. We present a review of the history of publications on the effects of environmental stressors on SCs, followed by a consolidation of the literature over the past five years on a subset of key environmental stressors of importance to human health and their effects on both embryonic and tissue SCs. The review will make constructive suggestions as to areas of toxicant research where further studies are needed, as well as making indications of the potential utility for advancing knowledge and directing research on environmental toxicology.
Collapse
Affiliation(s)
- Jessica R Worley
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI 48202, United States
| | - Graham C Parker
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI 48202, United States
| |
Collapse
|
16
|
Li L, Bi Z, Wadgaonkar P, Lu Y, Zhang Q, Fu Y, Thakur C, Wang L, Chen F. Metabolic and epigenetic reprogramming in the arsenic-induced cancer stem cells. Semin Cancer Biol 2019; 57:10-18. [PMID: 31009762 PMCID: PMC6690805 DOI: 10.1016/j.semcancer.2019.04.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 04/18/2019] [Indexed: 12/19/2022]
Abstract
At present, the belief that genetic mutations control every aspect of tumorigenesis is still very popular. Even for the highly debated "bad luck" theory of cancers, it ascertained that random mutation of genes during the self-renewal of somatic stem cells is responsible for cancer initiation. Logically, most of the new therapeutic strategies so far, from molecular targeting to precision medicine or personalized medicine, are genome-obsessed and focused on identifying and targeting these mutated genes. Accordingly, a rather simplified therapeutic regimen was formulated: cancers with the same mutations, e.g., lung cancer, pancreatic cancer, breast cancer, ovarian cancer, etc, were managed with the same chemo or targeting medicine, whereas for a particular cancer, such as breast cancer or lung cancer, with different mutational spectrums was treated with different, so-called personalized medicine. The outcomes of this strategy, however, are mixed with encouraging and disappointing findings. In this review article, we will address the importance of non-genetic factors, the metabolic and epigenetic reprogramming, during the induction of cancer stem cells in response to arsenic, a major environmental human carcinogen. The information provided may not only advance our understanding of carcinogenic mechanism to a new level but also help in designing new strategies through targeting the metabolic and epigenetic signaling pathways for cancer therapy.
Collapse
Affiliation(s)
- Lingzhi Li
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI, 48201, USA; Department of Developmental and Stem Cell Biology, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Zhuoyue Bi
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI, 48201, USA; School of Health Sciences, Wuhan University, No. 115, Donghu Road, Wuhan, 430071, Hubei, China; Hubei Provincial Key Laboratory of Applied Toxicology, Hubei Provincial Center for Disease Control and Prevention, 8 Zhuodaoquanbei Road, Wuhan, 430079, Hubei, China
| | - Priya Wadgaonkar
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI, 48201, USA
| | - Yongju Lu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI, 48201, USA
| | - Qian Zhang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI, 48201, USA
| | - Yao Fu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI, 48201, USA
| | - Chitra Thakur
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI, 48201, USA
| | - Li Wang
- Department of Physiology and Neurobiology and Institute for Systems Genomics, University of Connecticut, Storrs, CT, 06269, USA
| | - Fei Chen
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI, 48201, USA.
| |
Collapse
|
17
|
Sun K, Mei W, Mo S, Xin L, Lei X, Huang M, Chen Q, Han L, Zhu X. Lead exposure inhibits osteoblastic differentiation and inactivates the canonical Wnt signal and recovery by icaritin in MC3T3-E1 subclone 14 cells. Chem Biol Interact 2019; 303:7-13. [DOI: 10.1016/j.cbi.2019.01.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/20/2019] [Accepted: 01/31/2019] [Indexed: 10/27/2022]
|
18
|
Developmental neurotoxicity of inorganic arsenic exposure in Sprague-Dawley rats. Neurotoxicol Teratol 2019; 72:49-57. [DOI: 10.1016/j.ntt.2019.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/07/2018] [Accepted: 01/31/2019] [Indexed: 02/06/2023]
|
19
|
Sims KC, Schwendinger KL, Szymkowicz DB, Swetenburg JR, Bain LJ. Embryonic arsenic exposure reduces intestinal cell proliferation and alters hepatic IGF mRNA expression in killifish (Fundulus heteroclitus). JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2019; 82:142-156. [PMID: 30729860 PMCID: PMC6397093 DOI: 10.1080/15287394.2019.1571465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Arsenic (As) is a toxicant found in food and water throughout the world, and studies suggested that exposure early in life reduces growth. Thus, the goal of this study was to examine mechanisms by which As impacted organismal growth. Killifish (Fundulus heteroclitus) were exposed to 0, 10, 50, or 200 ppb As as embryos and, after hatching, were reared in clean water for up to 40 weeks. Metabolism studies revealed that killifish biotransform As such that monomethylated and dimethylated arsenicals account for 15-17% and 45-61%, respectively, of the total metal. Growth, as measured by condition factor (CF), was significantly and dose-dependently reduced at 8 weeks of age but was similar to controls by 40 weeks. To determine mechanisms underlying the observed initial decrease, intestinal proliferation and morphology were examined. Arsenic-exposed fish exhibited significant 1.3- to 1.5-fold reduction in intestinal villus height and 1.4- to 1.6-fold decrease in proliferating cell nuclear antigen (PCNA+) intestinal cells at all weeks examined. In addition, there were significant correlations between CF, PCNA+ cells, and intestinal villus height. Upon examining whether fish might compensate for the intestinal changes, it was found that hepatic mRNA expression of insulin-like growth factor 1 (IGF-1) and its binding protein (IGFBP-1) were dose-dependently increased. These results indicate that embryonic exposure initially diminished growth, and while intestinal cell proliferation remained reduced, fish appear to compensate by enhancing transcript levels of hepatic IGF-1 and IGFBP-1.
Collapse
Affiliation(s)
- Kaleigh C. Sims
- Environmental Toxicology Graduate Program, Clemson University, Clemson, SC, USA
| | | | - Dana B. Szymkowicz
- Environmental Toxicology Graduate Program, Clemson University, Clemson, SC, USA
| | | | - Lisa J. Bain
- Environmental Toxicology Graduate Program, Clemson University, Clemson, SC, USA
- Department of Biological Sciences, Clemson University, Clemson, SC, USA
| |
Collapse
|
20
|
Zhang Q, Bai J, Yao X, Jiang L, Wu W, Yang L, Gao N, Qiu T, Yang G, Habtemariam Hidru T, Sun X. Taurine rescues the arsenic-induced injury in the pancreas of rat offsprings and in the INS-1 cells. Biomed Pharmacother 2018; 109:815-822. [PMID: 30551535 DOI: 10.1016/j.biopha.2018.10.134] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 10/21/2018] [Accepted: 10/21/2018] [Indexed: 02/07/2023] Open
Abstract
Arsenic was an established carcinogen and toxicant, occurring in drinking water and food. Arsenic was increasingly being blamed as a risk factor for diabetes mellitus. Recent studies have found that arsenic could induce the generation of reactive oxygen species (ROS) and mitochondria were the major targets of ROS. Damage mitochondria could be removed by mitophagy and mitophagy played a defensive role against cellular apoptosis. To investigate whether the arsenic could induce the injury in mitochondria, we treated Wistar rat offsprings and INS-1 cells with As2O3 and sodium arsenite, respectively. Our results showed that arsenic induced the generation of ROS in both rat offsprings' pancreas and INS-1 cells. The generation of ROS induced by arsenic could inhibit the expression of PPARγ. PPARγ is a major impact on mitochondrial function. The inhibition of PPARγ induced the reduction of PINK1 signaling and the upregulation of Bax. PINK1 signaling was one of the classical pathways of mitophagy. The inhibition of mitophagy induced the activation of apoptosis both in rat offsprings' pancreas and INS-1 cells. After treated with Rosiglitazone (RGS, PPARγ receptor agonist), PPARγ was rescued, the expression of PINK1 significantly increasing and the apoptosis was restrained. We used Taurine (Tau) as the protective agent both in rat offsprings' pancreas and INS-1 cells, after treated with Tau, the production of ROS was decreased significantly and the downgrade of PPARγ was rescued.
Collapse
Affiliation(s)
- Qiaoting Zhang
- Department of Occupational and Environmental Health, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China
| | - Jie Bai
- Department of Nutrition and Food Hygiene, School of Public Health, Dalian medical university, 9W Lvshun South Road, Dalian 116044, PR China
| | - Xiaofeng Yao
- Department of Occupational and Environmental Health, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China
| | - Liping Jiang
- Department of Liaoning Anti-Degenerative Diseases Natural Products Engineering Research Center, Dalian Medical University, 9W Lvshun South Road, Dalian 116044, PR China
| | - Wei Wu
- Department of Occupational and Environmental Health, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China
| | - Lei Yang
- Department of Occupational and Environmental Health, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China
| | - Ni Gao
- Department of Occupational and Environmental Health, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China
| | - Tianming Qiu
- Department of Occupational and Environmental Health, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China
| | - Guang Yang
- Department of Liaoning Anti-Degenerative Diseases Natural Products Engineering Research Center, Dalian Medical University, 9W Lvshun South Road, Dalian 116044, PR China
| | | | - Xiance Sun
- Department of Occupational and Environmental Health, Dalian Medical University, 9 W Lvshun South Road, Dalian 116044, PR China.
| |
Collapse
|
21
|
Effect of subchronic exposure to inorganic arsenic on the structure and function of the intestinal epithelium. Toxicol Lett 2018; 286:80-88. [DOI: 10.1016/j.toxlet.2018.01.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/22/2017] [Accepted: 01/11/2018] [Indexed: 02/08/2023]
|
22
|
Liu JT, Bain LJ. Arsenic Induces Members of the mmu-miR-466-669 Cluster Which Reduces NeuroD1 Expression. Toxicol Sci 2018; 162:64-78. [PMID: 29121352 PMCID: PMC6693399 DOI: 10.1093/toxsci/kfx241] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Chronic arsenic exposure can result in adverse development effects including decreased intellectual function, reduced birth weight, and altered locomotor activity. Previous in vitro studies have shown that arsenic inhibits stem cell differentiation. MicroRNAs (miRNAs) are small noncoding RNAs that regulate multiple cellular processes including embryonic development and cell differentiation. The purpose of this study was to examine whether altered miRNA expression was a mechanism by which arsenic inhibited cellular differentiation. The pluripotent P19 mouse embryonal carcinoma cells were exposed to 0 or 0.5 μM sodium arsenite for 9 days during cell differentiation, and changes in miRNA expression was analyzed using microarrays. We found that the expression of several miRNAs important in cellular differentiation, such as miR-9 and miR-199 were decreased by 1.9- and 1.6-fold, respectively, following arsenic exposure, while miR-92a, miR-291a, and miR-709 were increased by 3-, 3.7-, and 1.6-fold, respectively. The members of the miR-466-669 cluster and its host gene, Scm-like with 4 Mbt domains 2 (Sfmbt2), were significantly induced by arsenic from 1.5- to 4-fold in a time-dependent manner. Multiple miRNA target prediction programs revealed that several neurogenic transcription factors appear to be targets of the cluster. When consensus anti-miRNAs targeting the miR-466-669 cluster were transfected into P19 cells, arsenic-exposed cells were able to more effectively differentiate. The consensus anti-miRNAs appeared to rescue the inhibitory effects of arsenic on cell differentiation due to an increased expression of NeuroD1. Taken together, we conclude that arsenic induces the miR-466-669 cluster, and that this induction acts to inhibit cellular differentiation in part due to a repression of NeuroD1.
Collapse
Affiliation(s)
| | - Lisa J Bain
- Environmental Toxicology Graduate Program
- Department of Biological Sciences, Clemson University, Clemson, South Carolina 29634
| |
Collapse
|
23
|
Margiotta AL, Bain LJ, Rice CD. Expression of the Major Vault Protein (MVP) and Cellular Vault Particles in Fish. Anat Rec (Hoboken) 2017; 300:1981-1992. [PMID: 28710803 DOI: 10.1002/ar.23645] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/04/2017] [Accepted: 07/06/2017] [Indexed: 12/15/2022]
Abstract
Cellular vaults are ubiquitous 13 mega Da multi-subunit ribonuceloprotein particles that may have a role in nucleocytoplasmic transport. Seventy percent of the vault's mass consists of a ≈100 kDa protein, the major vault protein (MVP). In humans, a drug resistance-associated protein, originally identified as lung resistance protein in metastatic lung cancer, was ultimately shown to be the previously described MVP. In this study, a partial MVP sequence was cloned from channel catfish. Recombinant MVP (rMVP) was used to generate a monoclonal antibody that recognizes full length protein in distantly related fish species, as well as mice. MVP is expressed in fish spleen, liver, anterior kidney, renal kidney, and gills, with a consistent expression in epithelial cells, macrophages, or endothelium at the interface of the tissue and environment or vasculature. We show that vaults are distributed throughout cells of fish lymphoid cells, with nuclear and plasma membrane aggregations in some cells. Protein expression studies were extended to liver neoplastic lesions in Atlantic killifish collected in situ at the Atlantic Wood USA-EPA superfund site on the southern branch of the Elizabeth River, VA. MVP is highly expressed in these lesions, with intense staining at the nuclear membrane, similar to what is known about MVP expression in human liver neoplasia. Additionally, MVP mRNA expression was quantified in channel catfish ovarian cell line following treatment with different classes of pharmacological agents. Notably, mRNA expression is induced by ethidium bromide, which damages DNA. Anat Rec, 2017. © 2017 Wiley Periodicals, Inc. Anat Rec, 300:1981-1992, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Alyssa L Margiotta
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, 29634
| | - Lisa J Bain
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, 29634.,Environmental Toxicology Graduate Program, Clemson University, Clemson, South Carolina, 29634
| | - Charles D Rice
- Department of Biological Sciences, Clemson University, Clemson, South Carolina, 29634.,Environmental Toxicology Graduate Program, Clemson University, Clemson, South Carolina, 29634
| |
Collapse
|