1
|
Amini A, Esmaeili F, Golpich M. Possible role of lncRNAs in amelioration of Parkinson's disease symptoms by transplantation of dopaminergic cells. NPJ Parkinsons Dis 2024; 10:56. [PMID: 38472261 PMCID: PMC10933336 DOI: 10.1038/s41531-024-00661-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/14/2024] [Indexed: 03/14/2024] Open
Abstract
Long non-coding RNAs (lncRNAs) are biomarkers for diagnosis and treatment of Parkinson's disease (PD). Since dopaminergic cell transplantation is a clinical method to treat PD, this study investigated the effects of dopaminergic cell therapy on the expression of some lncRNAs and genes related to PD. In this study, Twenty-eight rats were randomly assigned to four experimental groups. The control group (Sal group) received saline injections. The Par group was a PD rat model with 6-hydroxydopamine (6-OHDA) injection in right striatum (ST). PD animals were transplanted by undifferentiated P19 stem cells (Par-E group), and P19-derived dopaminergic cells (Par-N group). Cell transplant effects were evaluated using behavioral tests (cylinder, open field, and rotarod tests), and histological methods (H&E and Nissl staining, and immunohistochemistry). Moreover, the expression of lncRNAs MALAT1, MEG3, and SNHG1, alongside specific neuronal (synaptophysin) and dopaminergic (tyrosine hydroxylase) markers was evaluated by qRT-PCR. Behavioral and histopathological examinations revealed that cell transplantation partially compensated dopaminergic cell degeneration in ST and substantia nigra (SN) of PD rats. The expression of MALAT1, SNHG1, and MEG3 was decreased in the ST of the Par group, while MEG3 and SNHG1 gene expression was increased in PBMC relative to the Sal group. In PBMC of the Par-N group, all three lncRNAs showed a reduction in their expression. Conversely, MALAT1 and SNHG1 expression was increased in ST tissue, while MEG3 gene expression was decreased compared to the Sal group. In conclusion, dopaminergic cell transplantation could change the lncRNAs expression. Furthermore, it partially improves symptoms in PD rats.
Collapse
Affiliation(s)
- A Amini
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - F Esmaeili
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - M Golpich
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
2
|
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: 0] [Impact Index Per Article: 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
|
3
|
Lashani E, Amoozegar MA, Turner RJ, Moghimi H. Use of Microbial Consortia in Bioremediation of Metalloid Polluted Environments. Microorganisms 2023; 11:microorganisms11040891. [PMID: 37110315 PMCID: PMC10143001 DOI: 10.3390/microorganisms11040891] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 03/31/2023] Open
Abstract
Metalloids are released into the environment due to the erosion of the rocks or anthropogenic activities, causing problems for human health in different world regions. Meanwhile, microorganisms with different mechanisms to tolerate and detoxify metalloid contaminants have an essential role in reducing risks. In this review, we first define metalloids and bioremediation methods and examine the ecology and biodiversity of microorganisms in areas contaminated with these metalloids. Then we studied the genes and proteins involved in the tolerance, transport, uptake, and reduction of these metalloids. Most of these studies focused on a single metalloid and co-contamination of multiple pollutants were poorly discussed in the literature. Furthermore, microbial communication within consortia was rarely explored. Finally, we summarized the microbial relationships between microorganisms in consortia and biofilms to remove one or more contaminants. Therefore, this review article contains valuable information about microbial consortia and their mechanisms in the bioremediation of metalloids.
Collapse
Affiliation(s)
- Elham Lashani
- Extremophiles Laboratory, Department of Microbiology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran 14178-64411, Iran;
| | - Mohammad Ali Amoozegar
- Extremophiles Laboratory, Department of Microbiology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran 14178-64411, Iran;
- Correspondence: (M.A.A.); (H.M.); Tel.: +98-21-66415495 (H.M.)
| | - Raymond J. Turner
- Microbial Biochemistry Laboratory, Department of Biological Sciences, University of Calgary, 2500 University Dr. NW, Calgary, AB T2N 1N4, Canada;
| | - Hamid Moghimi
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran 14178-64411, Iran
- Correspondence: (M.A.A.); (H.M.); Tel.: +98-21-66415495 (H.M.)
| |
Collapse
|
4
|
McMichael BD, Perego MC, Darling CL, Perry RL, Coleman SC, Bain LJ. Long-term arsenic exposure impairs differentiation in mouse embryonal stem cells. J Appl Toxicol 2020; 41:1089-1102. [PMID: 33124703 DOI: 10.1002/jat.4095] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 11/12/2022]
Abstract
Arsenic is a contaminant found in many foods and drinking water. Exposure to arsenic during development can cause improper neuronal progenitor cell development, differentiation, and function, while in vitro studies have determined that acute arsenic exposure to stem and progenitor cells reduced their ability to differentiate. In the current study, P19 mouse embryonal stem cells were exposed continuously to 0.1-μM (7.5 ppb) arsenic for 32 weeks. A cell lineage array examining messenger RNA (mRNA) changes after 8 and 32 weeks of exposure showed that genes involved in pluripotency were increased, whereas those involved in differentiation were reduced. Therefore, temporal changes of select pluripotency and neuronal differentiation markers throughout the 32-week chronic arsenic exposure were investigated. Sox2 and Oct4 mRNA expression were increased by 1.9- to 2.5-fold in the arsenic-exposed cells, beginning at Week 12. Sox2 protein expression was similarly increased starting at Week 16 and remained elevated by 1.5-fold to sixfold. One target of Sox2 is N-cadherin, whose expression is a hallmark of epithelial-mesenchymal transitions (EMTs). Exposure to arsenic significantly increased N-cadherin protein levels beginning at Week 20, concurrent with increased grouping of N-cadherin positive cells at the perimeter of the embryoid body. Expression of Zeb1, which helps increase the expression of Sox2, was also increased started at Week 16. In contrast, Gdf3 mRNA expression was reduced by 3.4- to 7.2-fold beginning at Week 16, and expression of its target protein, phospho-Smad2/3, was also reduced. These results suggest that chronic, low-level arsenic exposure may delay neuronal differentiation and maintain pluripotency.
Collapse
Affiliation(s)
- Benjamin D McMichael
- Environmental Toxicology Graduate Program, Clemson University, Clemson, South Carolina, USA.,US Environmental Protection Agency, Durham, North Carolina, USA
| | - M Chiara Perego
- Environmental Toxicology Graduate Program, Clemson University, Clemson, South Carolina, USA
| | - Caitlin L Darling
- Environmental Toxicology Graduate Program, Clemson University, Clemson, South Carolina, USA
| | - Rebekah L Perry
- Environmental Toxicology Graduate Program, Clemson University, Clemson, South Carolina, USA
| | - Sarah C Coleman
- Environmental Toxicology Graduate Program, Clemson University, Clemson, South Carolina, USA
| | - Lisa J Bain
- Environmental Toxicology Graduate Program, Clemson University, Clemson, South Carolina, USA.,Department of Biological Sciences, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
5
|
Sanyal T, Bhattacharjee P, Paul S, Bhattacharjee P. Recent Advances in Arsenic Research: Significance of Differential Susceptibility and Sustainable Strategies for Mitigation. Front Public Health 2020; 8:464. [PMID: 33134234 PMCID: PMC7578365 DOI: 10.3389/fpubh.2020.00464] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/23/2020] [Indexed: 12/16/2022] Open
Abstract
Arsenic contamination in drinking water and associated adverse outcomes are one of the major health issues in more than 50 countries worldwide. The scenario is getting even more detrimental with increasing number of affected people and newer sites reported from all over the world. Apart from drinking water, the presence of arsenic has been found in various other dietary sources. Chronic arsenic toxicity affects multiple physiological systems and may cause malignancies leading to death. Exposed individuals, residing in the same area, developed differential dermatological lesion phenotypes and varied susceptibility toward various other arsenic-induced disease risk, even after consuming equivalent amount of arsenic from the similar source, over the same duration of time. Researches so far indicate that differential susceptibility plays an important role in arsenic-induced disease manifestation. In this comprehensive review, we have identified major population-based studies of the last 20 years, indicating possible causes of differential susceptibility emphasizing arsenic methylation capacity, variation in host genome (single nucleotide polymorphism), and individual epigenetic pattern (DNA methylation, histone modification, and miRNA expression). Holistic multidisciplinary strategies need to be implemented with few sustainable yet cost-effective solutions like alternative water source, treatment of arsenic-contaminated water, new adaptations in irrigation system, simple modifications in cooking strategy, and dietary supplementations to combat this menace. Our review focuses on the present perspectives of arsenic research with special emphasis on the probable causes of differential susceptibility toward chronic arsenic toxicity and sustainable remediation strategies.
Collapse
Affiliation(s)
- Tamalika Sanyal
- Department of Zoology, University of Calcutta, Kolkata, India.,Department of Environmental Science, University of Calcutta, Kolkata, India
| | - Pritha Bhattacharjee
- Department of Zoology, University of Calcutta, Kolkata, India.,Department of Environmental Science, University of Calcutta, Kolkata, India
| | - Somnath Paul
- Department of Epigenetics and Molecular Carcinogenesis, U.T. MD Anderson Cancer Center, Smithville, TX, United States
| | | |
Collapse
|
6
|
Qin Y, Jiang M, Tuerxung N, Wang H, Zhao F, Zhen Y, Hao J. Sonic hedgehog signaling pathway in Myelodysplastic Syndrome: Abnormal activation and jervine intervention. Gene 2020; 754:144881. [PMID: 32526259 DOI: 10.1016/j.gene.2020.144881] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/27/2020] [Accepted: 06/06/2020] [Indexed: 01/30/2023]
Abstract
OBJECTIVE This study aims to investigate the roles of Sonic hedgehog (Shh) signaling pathway in the occurrence and progression of Myelodysplastic Syndrome (MDS) and further evaluate using jervine as therapeutic strategy for MDS by inhibiting Shh pathway. METHODS CD34+ cells from the bone marrow of 53 MDS patients were counted by flow cytometry and isolated by magnetic bead sorting. Shh, Smo, Ptch-1 and Gli-1 (involved in Shh pathway) in CD34+ cells were examined by RT-qPCR. Besides, the relationship between Shh pathway-related genes and the clinical features or prognosis of MDS were analyzed. Further, the effects of jervine on MUTZ-1 cells regarding their proliferation, apoptosis and cell cycle as well as Shh pathway-related gene and protein expression were analyzed. RESULTS Gene expression level of Shh, Gli-1 and Smo was significantly increased in MDS patients. Herein, Smo and Gli-1 were correlated with chromosome karyotype classification and IPSS. MDS patients with high expression of Smo or Gli-1 had a poor prognosis. Jervine inhibited gene and protein expression of Shh, Smo, Ptch-1 and Gli-1. Besides, jervine suppressed the proliferation and promoted the apoptosis of MUTZ-1 cells, as well as inhibited the transition of cells from G1 to S phase. CONCLUSION Shh signaling pathway of MDS patients is abnormally activated and participated in the occurrence and progression of MDS. Jervine intervention is a potential therapeutic strategy for MDS.
Collapse
Affiliation(s)
- YuTing Qin
- Hematologic Disease Center, The First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Xinshi District, Urumqi, Xinjiang Province 830054, China
| | - Ming Jiang
- Hematologic Disease Center, The First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Xinshi District, Urumqi, Xinjiang Province 830054, China
| | - Nilupar Tuerxung
- Hematologic Disease Center, The First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Xinshi District, Urumqi, Xinjiang Province 830054, China
| | - Huan Wang
- Hematologic Disease Center, The First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Xinshi District, Urumqi, Xinjiang Province 830054, China
| | - Fang Zhao
- Hematologic Disease Center, The First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Xinshi District, Urumqi, Xinjiang Province 830054, China
| | - Yin Zhen
- Hematologic Disease Center, The First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Xinshi District, Urumqi, Xinjiang Province 830054, China
| | - Jianping Hao
- Hematologic Disease Center, The First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan Road, Xinshi District, Urumqi, Xinjiang Province 830054, China.
| |
Collapse
|
7
|
Sanyal T, Bhattacharjee P, Paul S, Bhattacharjee P. Recent Advances in Arsenic Research: Significance of Differential Susceptibility and Sustainable Strategies for Mitigation. Front Public Health 2020. [PMID: 33134234 DOI: 10.3389/fpubh/2020.00464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023] Open
Abstract
Arsenic contamination in drinking water and associated adverse outcomes are one of the major health issues in more than 50 countries worldwide. The scenario is getting even more detrimental with increasing number of affected people and newer sites reported from all over the world. Apart from drinking water, the presence of arsenic has been found in various other dietary sources. Chronic arsenic toxicity affects multiple physiological systems and may cause malignancies leading to death. Exposed individuals, residing in the same area, developed differential dermatological lesion phenotypes and varied susceptibility toward various other arsenic-induced disease risk, even after consuming equivalent amount of arsenic from the similar source, over the same duration of time. Researches so far indicate that differential susceptibility plays an important role in arsenic-induced disease manifestation. In this comprehensive review, we have identified major population-based studies of the last 20 years, indicating possible causes of differential susceptibility emphasizing arsenic methylation capacity, variation in host genome (single nucleotide polymorphism), and individual epigenetic pattern (DNA methylation, histone modification, and miRNA expression). Holistic multidisciplinary strategies need to be implemented with few sustainable yet cost-effective solutions like alternative water source, treatment of arsenic-contaminated water, new adaptations in irrigation system, simple modifications in cooking strategy, and dietary supplementations to combat this menace. Our review focuses on the present perspectives of arsenic research with special emphasis on the probable causes of differential susceptibility toward chronic arsenic toxicity and sustainable remediation strategies.
Collapse
Affiliation(s)
- Tamalika Sanyal
- Department of Zoology, University of Calcutta, Kolkata, India
- Department of Environmental Science, University of Calcutta, Kolkata, India
| | - Pritha Bhattacharjee
- Department of Zoology, University of Calcutta, Kolkata, India
- Department of Environmental Science, University of Calcutta, Kolkata, India
| | - Somnath Paul
- Department of Epigenetics and Molecular Carcinogenesis, U.T. MD Anderson Cancer Center, Smithville, TX, United States
| | | |
Collapse
|
8
|
Nam A, Kim T, Li Q, Rebhun RB, Youn HY, Seo KW. Melarsomine suppresses canine osteosarcoma cell survival via inhibition of Hedgehog-GLI signaling. J Vet Med Sci 2019; 81:1722-1729. [PMID: 31645504 PMCID: PMC6943322 DOI: 10.1292/jvms.19-0043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The Hedgehog-GLI signaling pathway is activated in human and canine osteosarcoma (OSA)
and represents a potential therapeutic target for cancers, including OSA. Arsenic trioxide
represses GLI expression. Melarsomine, an arsenic compound-containing drug, has been
approved for the treatment of canine heartworm disease. Hence, we hypothesized that
melarsomine inhibits GLI signaling in canine OSA cell lines. The present study aimed to
assess this hypothesis. Cell viability and colony formation were decreased in the canine
OSA cell lines Abrams and D17 after treatment with melarsomine. Melarsomine-induced
apoptotic cell death was assessed via cell cycle analysis using propidium iodide staining.
Quantitative real-time reverse transcription polymerase chain reaction and western blot
analyses revealed a downregulation of genes downstream of the Hedgehog signaling pathway,
including GLI1, GLI2, and PTCH, after
melarsomine treatment. The present results suggest that melarsomine exerts antitumor
effects and serves as a GLI inhibitor in canine OSA cells. Additional studies are required
to evaluate and confirm the anticancer effect and relevant therapeutic dose of melarsomine
in vivo.
Collapse
Affiliation(s)
- Aryung Nam
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Taewon Kim
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, South Korea
| | - Qiang Li
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Robert B Rebhun
- Department of Surgical and Radiological Sciences, University of California Davis, School of Veterinary Medicine, Davis 95616, CA, U.S.A
| | - Hwa-Young Youn
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Kyoung-Won Seo
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|
9
|
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: 6.2] [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
|
10
|
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: 0.4] [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
|
11
|
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: 1.3] [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
|
12
|
Beeravolu N, McKee C, Chaudhry GR. Mechanism of arsenite toxicity in embryonic stem cells. J Appl Toxicol 2017; 37:1151-1161. [PMID: 28370166 DOI: 10.1002/jat.3469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 01/31/2017] [Accepted: 02/21/2017] [Indexed: 11/11/2022]
Abstract
Environmental arsenite exposure has been linked to cancer as well as other diseases, presenting an important and serious public health problem. Toxicity of inorganic arsenite (iAs) has been investigated using animal models and cell culture, yet its developmental effects are poorly understood. This study investigated the molecular mechanism of iAs toxicity to ascertain insight into development and differentiation processes using mouse embryonic stem cells (ESCs). The results showed that iAs exposure affected morphology and integrity of ESC colonies as well as inhibited cell growth in a concentration-dependent manner, excluding concentrations <1 μM iAs which stimulated ESC growth. ESCs self-renewal and pluripotency was also affected as evident from the downregulation of transcription circuitry, Oct4, Nanog, Sox2 and Klf4 resulting in non-specific differentiation. ESCs exposed to iAs randomly differentiated into three germ layers, mesoderm, endoderm and ectoderm, as judged by transcriptional expression of Brachyury, Gata4 and FGF2, as well as translational expression of BRACHYURY, GATA4 and TUJ1 respectively. The differentiated cells represented osteogenic, chondrogenic, myogenic and neurogenic lineages as evident from upregulation of Col1, Sox9, Col2, Myog, Notch, Nes and Nef. Although iAs caused slight apoptosis with a concomitant increase in ROS levels, the exposed ESCs had significant Bcl2 expression, which could be involved in the protection against apoptosis. Further analysis revealed upregulation of Jun and P38 in ESCs with an increase in iAs concentration. These observations indicated that iAs stress caused random differentiation of ESCs via JNK/P38 pathways. These findings suggest that iAs exposure may cause teratogenicity during early fetal development. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Naimisha Beeravolu
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA.,OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, Michigan, USA
| | - Christina McKee
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA.,OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, Michigan, USA
| | - G Rasul Chaudhry
- Department of Biological Sciences, Oakland University, Rochester, Michigan, USA.,OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, Michigan, USA
| |
Collapse
|
13
|
Tyler CR, Labrecque MT, Solomon ER, Guo X, Allan AM. Prenatal arsenic exposure alters REST/NRSF and microRNA regulators of embryonic neural stem cell fate in a sex-dependent manner. Neurotoxicol Teratol 2016; 59:1-15. [PMID: 27751817 DOI: 10.1016/j.ntt.2016.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 08/30/2016] [Accepted: 10/13/2016] [Indexed: 11/29/2022]
Abstract
Exposure to arsenic, a common environmental toxin found in drinking water, leads to a host of neurological pathologies. We have previously demonstrated that developmental exposure to a low level of arsenic (50ppb) alters epigenetic processes that underlie deficits in adult hippocampal neurogenesis leading to aberrant behavior. It is unclear if arsenic impacts the programming and regulation of embryonic neurogenesis during development when exposure occurs. The master negative regulator of neural-lineage, REST/NRSF, controls the precise timing of fate specification and differentiation of neural stem cells (NSCs). Early in development (embryonic day 14), we observed increased expression of Rest, its co-repressor, CoREST, and the inhibitory RNA binding/splicing protein, Ptbp1, and altered expression of mRNA spliced isoforms of Pbx1 that are directly regulated by these factors in the male brain in response to prenatal 50ppb arsenic exposure. These increases were concurrent with decreased expression of microRNA-9 (miR-9), miR-9*, and miR-124, all of which are REST/NRSF targets and inversely regulate Rest expression to allow for maturation of NSCs. Exposure to arsenic decreased the formation of neuroblasts in vitro from NSCs derived from male pup brains. The female response to arsenic was limited to increased expression of CoREST and Ptbp2, an RNA binding protein that allows for appropriate splicing of genes involved in the progression of neurogenesis. These changes were accompanied by increased neuroblast formation in vitro from NSCs derived from female pups. Unexposed male mice express transcriptomic factors to induce differentiation earlier in development compared to unexposed females. Thus, arsenic exposure likely delays differentiation of NSCs in males while potentially inducing precocious differentiation in females early in development. These effects are mitigated by embryonic day 18 of development. Arsenic-induced dysregulation of the regulatory loop formed by REST/NRSF, its target microRNAs, miR-9 and miR-124, and RNA splicing proteins, PTBP1 and 2, leads to aberrant programming of NSC function that is perhaps perpetuated into adulthood inducing deficits in differentiation we have previously observed.
Collapse
Affiliation(s)
- Christina R Tyler
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM 87131, United States
| | - Matthew T Labrecque
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, United States
| | - Elizabeth R Solomon
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, United States
| | - Xun Guo
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, United States
| | - Andrea M Allan
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, United States.
| |
Collapse
|
14
|
He YH, Li Z, Ni MM, Zhang XY, Li MF, Meng XM, Huang C, Li J. Cryptolepine derivative-6h inhibits liver fibrosis in TGF-β1-induced HSC-T6 cells by targeting the Shh pathway. Can J Physiol Pharmacol 2016; 94:987-95. [PMID: 27295431 DOI: 10.1139/cjpp-2016-0157] [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] [Indexed: 12/21/2022]
Abstract
Liver fibrosis is a worldwide problem with a significant morbidity and mortality. Cryptolepis sanguinolenta (family Periplocaceae) is widely used in West African countries for the treatment of malaria, as well as for some other diseases. However, the role of C. sanguinolenta in hepatic fibrosis is still unknown. It has been reported that Methyl-CpG binding protein 2 (MeCP2) had a high expression in liver fibrosis and played a central role in its pathobiology. Interestingly, we found that a cryptolepine derivative (HZ-6h) could inhibit liver fibrosis by reducing MeCP2 expression, as evidenced by the dramatic downregulation of α-smooth muscle actin (α-SMA) and type I collagen alpha-1 (Col1α1) in protein levels in vitro. Meanwhile, we also found that HZ-6h could reduce the cell viability and promote apoptosis of hepatic stellate cells (HSCs) treated with transforming growth factor beta 1(TGF-β1). Then, we investigated the potential molecular mechanisms and found that HZ-6h blocked Shh signaling in HSC-T6 cells, resulting in the decreased protein expression of Patched-1 (PTCH-1), Sonic hedgehog (Shh), and glioma-associated oncogene homolog 1 (GLI1). In short, these results indicate that HZ-6h inhibits liver fibrosis by downregulating MeCP2 through the Shh pathway in TGF-β1-induced HSC-T6 cells.
Collapse
Affiliation(s)
- Ying-Hua He
- a School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China.,b Institute for Liver Diseases, Anhui Medical University, ILD-AMU, Hefei 230032, China.,c Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China
| | - Zeng Li
- a School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China.,b Institute for Liver Diseases, Anhui Medical University, ILD-AMU, Hefei 230032, China.,c Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China
| | - Ming-Ming Ni
- a School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China.,b Institute for Liver Diseases, Anhui Medical University, ILD-AMU, Hefei 230032, China.,c Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China
| | - Xing-Yan Zhang
- a School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China.,b Institute for Liver Diseases, Anhui Medical University, ILD-AMU, Hefei 230032, China.,c Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China
| | - Ming-Fang Li
- a School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China.,b Institute for Liver Diseases, Anhui Medical University, ILD-AMU, Hefei 230032, China.,c Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China
| | - Xiao-Ming Meng
- a School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China.,b Institute for Liver Diseases, Anhui Medical University, ILD-AMU, Hefei 230032, China.,c Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China
| | - Cheng Huang
- a School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China.,b Institute for Liver Diseases, Anhui Medical University, ILD-AMU, Hefei 230032, China.,c Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China
| | - Jun Li
- a School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China.,b Institute for Liver Diseases, Anhui Medical University, ILD-AMU, Hefei 230032, China.,c Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
15
|
Bain LJ, Liu JT, League RE. Arsenic inhibits stem cell differentiation by altering the interplay between the Wnt3a and Notch signaling pathways. Toxicol Rep 2016; 3:405-413. [PMID: 27158593 PMCID: PMC4855706 DOI: 10.1016/j.toxrep.2016.03.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
data indicates that arsenic exposure inhibits stem cell differentiation. This study investigated whether arsenic disrupted the Wnt3a signaling pathway, critical in the formation of myotubes and neurons, during the differentiation in P19 mouse embryonic stem cells. Cells were exposed to 0, 0.1, or 0.5 μM arsenite, with or without exogenous Wnt3a, for up to 9 days of differentiation. Arsenic exposure alone inhibits the differentiation of stem cells into neurons and skeletal myotubes, and reduces the expression of both β-catenin and GSK3β mRNA to ~55% of control levels. Co-culture of the arsenic-exposed cells with exogenous Wnt3a rescues the morphological phenotype, but does not alter transcript, protein, or phosphorylation status of GSK3β or β-catenin. However, arsenic exposure maintains high levels of Hes5 and decreases the expression of MASH1 by 2.2-fold, which are anti- and pro-myogenic and neurogenic genes, respectively, in the Notch signaling pathway. While rescue with exogenous Wnt3a reduced Hes5 levels, MASH1 levels stay repressed. Thus, while Wnt3a can partially rescue the inhibition of differentiation from arsenic, it does so by also modulating Notch target genes rather than only working through the canonical Wnt signaling pathway. These results indicate that arsenic alters the interplay between multiple signaling pathways, leading to reduced stem cell differentiation.
Collapse
Affiliation(s)
- 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 23964, USA
| | - Jui-Tung Liu
- Environmental Toxicology Graduate Program, Clemson University, 132 Long Hall, Clemson, SC 29634, USA
| | - Ryan E League
- Environmental Toxicology Graduate Program, Clemson University, 132 Long Hall, Clemson, SC 29634, USA
| |
Collapse
|
16
|
Wu X, Zhao B, Cheng Y, Yang Y, Huang C, Meng X, Wu B, Zhang L, Lv X, Li J. Melittin induces PTCH1 expression by down-regulating MeCP2 in human hepatocellular carcinoma SMMC-7721 cells. Toxicol Appl Pharmacol 2015; 288:74-83. [PMID: 26189965 DOI: 10.1016/j.taap.2015.07.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 06/29/2015] [Accepted: 07/15/2015] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) has a high mortality rate worldwide and still remains to be a noticeable public health problem. Therefore, new remedies are urgently needed. Melittin, a major component of bee venom, is known to suppress cell growth in various cancers including HCC. However, the mechanism of the anticancer effect of melittin on HCC has not been fully elucidated. It has been reported that Methyl-CpG binding protein 2 (MeCP2) plays a key role in tumor proliferation, apoptosis, migration and invasion. In the present study, we found the high expression of MeCP2 in human HCC tissues and in the SMMC-7721 cell line. MeCP2 silencing inhibited cell proliferation, while over-expression of MeCP2 promoted cell growth in SMMC-7721 cells. It indicates that MeCP2 may be an attractive target for human HCC. We further found that melittin could inhibit cell proliferation by reducing MeCP2 expression in vitro. Interestingly, the inhibitory effect of melittin on cell proliferation was due to a delay in G0/G1 cell cycle progression, without influencing cell apoptosis. Next, we investigated the potential molecular mechanisms and found that MeCP2 could modulate Shh signaling in SMMC-7721 cells. Further study indicates that melittin may induce the demethylation of PTCH1 promoter, resulting in the increased expression of PTCH1. Furthermore, the expression of Shh and GLI1 was significantly lowered upon treatment of melittin. These results suggest that melittin can block Shh signaling in vitro. In short, these results indicate that melittin inhibits cell proliferation by down-regulating MeCP2 through Shh signaling in SMMC-7721 cells.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Cycle Checkpoints/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- DNA Methylation
- Dose-Response Relationship, Drug
- Gene Expression Regulation, Neoplastic
- Hedgehog Proteins/metabolism
- Humans
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Melitten/pharmacology
- Methyl-CpG-Binding Protein 2/genetics
- Methyl-CpG-Binding Protein 2/metabolism
- Patched Receptors
- Patched-1 Receptor
- Promoter Regions, Genetic
- RNA Interference
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Resting Phase, Cell Cycle/drug effects
- Signal Transduction/drug effects
- Time Factors
- Transcription Factors/metabolism
- Transfection
- Zinc Finger Protein GLI1
Collapse
Affiliation(s)
- Xiaoqin Wu
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, ILD-AMU, Anhui Medical University, Hefei 230032, China
| | - Bin Zhao
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, ILD-AMU, Anhui Medical University, Hefei 230032, China
| | - Yahui Cheng
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, ILD-AMU, Anhui Medical University, Hefei 230032, China
| | - Yang Yang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, ILD-AMU, Anhui Medical University, Hefei 230032, China
| | - Cheng Huang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, ILD-AMU, Anhui Medical University, Hefei 230032, China
| | - Xiaoming Meng
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, ILD-AMU, Anhui Medical University, Hefei 230032, China
| | - Baoming Wu
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, ILD-AMU, Anhui Medical University, Hefei 230032, China
| | - Lei Zhang
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, ILD-AMU, Anhui Medical University, Hefei 230032, China
| | - Xiongwen Lv
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, ILD-AMU, Anhui Medical University, Hefei 230032, China
| | - Jun Li
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, ILD-AMU, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
17
|
McCoy CR, Stadelman BS, Brumaghim JL, Liu JT, Bain LJ. Arsenic and Its Methylated Metabolites Inhibit the Differentiation of Neural Plate Border Specifier Cells. Chem Res Toxicol 2015; 28:1409-21. [PMID: 26024302 DOI: 10.1021/acs.chemrestox.5b00036] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Exposure to arsenic in food and drinking water has been correlated with adverse developmental outcomes, such as reductions in birth weight and neurological deficits. Additionally, studies have shown that arsenic suppresses sensory neuron formation and skeletal muscle myogenesis, although the reason why arsenic targets both of these cell types in unclear. Thus, P19 mouse embryonic stem cells were used to investigate the mechanisms by which arsenic could inhibit cellular differentiation. P19 cells were exposed to 0, 0.1, or 0.5 μM sodium arsenite and induced to form embryoid bodies over a period of 5 days. The expression of transcription factors necessary to form neural plate border specifier (NPBS) cells, neural crest cells and their progenitors, and myocytes and their progenitors were examined. Early during differentiation, arsenic significantly reduced the transcript and protein expression of Msx1 and Pax3, both needed for NPBS cell formation. Arsenic also significantly reduced the protein expression of Sox 10, needed for neural crest progenitor cell production, by 31-50%, and downregulated the protein and mRNA levels of NeuroD1, needed for neural crest cell differentiation, in a time- and dose-dependent manner. While the overall protein expression of transcription factors in the skeletal muscle lineage was not changed, arsenic did alter their nuclear localization. MyoD nuclear translocation was significantly reduced on days 2-5 between 15 and 70%. At a 10-fold lower concentration, monomethylarsonous acid (MMA III) appeared to be just as potent as inorganic arsenic at reducing the mRNA levels Pax3 (79% vs84%), Sox10 (49% vs 65%), and Msx1 (56% vs 56%). Dimethylarsinous acid (DMA III) also reduced protein and transcript expression, but the changes were less dramatic than those with MMA or arsenite. All three arsenic species reduced the nuclear localization of MyoD and NeuroD1 in a similar manner. The early changes in the differentiation of neural plate border specifier cells may provide a mechanism for arsenic to suppress both neurogenesis and myogenesis.
Collapse
Affiliation(s)
- Christopher R McCoy
- †Department of Biological Sciences, Clemson University, 132 Long Hall, Clemson, South Carolina 29634, United States
| | - Bradley S Stadelman
- ‡Department of Chemistry, Clemson University, 219 Hunter Laboratories, Clemson, South Carolina 29634, United States
| | - Julia L Brumaghim
- ‡Department of Chemistry, Clemson University, 219 Hunter Laboratories, Clemson, South Carolina 29634, United States
| | - Jui-Tung Liu
- §Environmental Toxicology Graduate Program, Clemson University, 132 Long Hall, Clemson, South Carolina 29634, United States
| | - Lisa J Bain
- †Department of Biological Sciences, Clemson University, 132 Long Hall, Clemson, South Carolina 29634, United States.,§Environmental Toxicology Graduate Program, Clemson University, 132 Long Hall, Clemson, South Carolina 29634, United States
| |
Collapse
|
18
|
Aung KH, Tsukahara S, Maekawa F, Nohara K, Nakamura K, Tanoue A. Role of Environmental Chemical Insult in Neuronal Cell Death and Cytoskeleton Damage. Biol Pharm Bull 2015; 38:1109-12. [DOI: 10.1248/bpb.b14-00890] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Kyaw Htet Aung
- Department of Pharmacology, National Research Institute for Child Health and Development
- Division of Life Science, Graduate School of Science and Engineering, Saitama University
| | - Shinji Tsukahara
- Division of Life Science, Graduate School of Science and Engineering, Saitama University
| | - Fumihiko Maekawa
- Molecular Toxicology Section, Center for Environmental Health Sciences, National Institute for Environmental Studies
| | - Keiko Nohara
- Molecular Toxicology Section, Center for Environmental Health Sciences, National Institute for Environmental Studies
| | - Kazuaki Nakamura
- Department of Pharmacology, National Research Institute for Child Health and Development
| | - Akito Tanoue
- Department of Pharmacology, National Research Institute for Child Health and Development
| |
Collapse
|