1
|
Li X, Li G, Cui S, Hou Y, Li Z, Yan Z, Huang T, Zhao T, Su H, Zhou B, Zhang J, Ao R, Zhao H, Qiu Y, Liu Z, Xie J. Arsenic disturbs neural tube closure involving AMPK/PKB-mTORC1-mediated autophagy in mice. Food Chem Toxicol 2024; 186:114538. [PMID: 38387523 DOI: 10.1016/j.fct.2024.114538] [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: 12/04/2023] [Revised: 02/01/2024] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
Arsenic exposure is a significant risk factor for folate-resistant neural tube defects (NTDs), but the potential mechanism is unclear. In this study, a mouse model of arsenic-induced NTDs was established to investigate how arsenic affects early neurogenesis leading to malformations. The results showed that in utero exposure to arsenic caused a decline in the normal embryos, an elevated embryo resorption, and a higher incidence of malformed embryos. Cranial and spinal deformities were the main malformation phenotypes observed. Meanwhile, arsenic-induced NTDs were accompanied by an oxidant/antioxidant imbalance manifested by elevated levels of reactive oxygen species (ROS) and decreased antioxidant activities. In addition, changes in the expression of autophagy-related genes and proteins (ULK1, Atg5, LC3B, p62) as well as an increase in autophagosomes were observed in arsenic-induced aberrant brain vesicles. Also, the components of the upstream pathway regulating autophagy (AMPK, PKB, mTOR, Raptor) were altered accordingly after arsenic exposure. Collectively, our findings propose a mechanism for arsenic-induced NTDs involving AMPK/PKB-mTORC1-mediated autophagy. Blocking autophagic cell death due to excessive autophagy provides a novel strategy for the prevention of folate-resistant NTDs, especially for arsenic-exposed populations.
Collapse
Affiliation(s)
- Xiujuan Li
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, 030001, China
| | - Gexuan Li
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, 030001, China; School of Public Health, Shanxi Medical University, Taiyuan, 030001, China
| | - Shuo Cui
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, 030001, China; School of Public Health, Shanxi Medical University, Taiyuan, 030001, China
| | - Yue Hou
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, 030001, China; School of Public Health, Shanxi Medical University, Taiyuan, 030001, China
| | - Zelin Li
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, 030001, China
| | - Ziyi Yan
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, 030001, China; School of Public Health, Shanxi Medical University, Taiyuan, 030001, China
| | - Tingjuan Huang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, 030001, China
| | - Taoran Zhao
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, 030001, China
| | - Hongkai Su
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, 030001, China
| | - Bingrui Zhou
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, 030001, China
| | - Juan Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, 030001, China
| | - Ruifang Ao
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, 030001, China
| | - Hong Zhao
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, 030001, China
| | - Yulan Qiu
- School of Public Health, Shanxi Medical University, Taiyuan, 030001, China
| | - Zhizhen Liu
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, 030001, China.
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
2
|
Xu FX, Chen X, Zhang H, Fan YJ, Song YP, Lv JW, Xie YL, Huang Y, Chen DZ, Wang H, Xu DX. Association between gestational arsenic exposure and intrauterine growth restriction: the role of folate content. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:89652-89661. [PMID: 35857162 DOI: 10.1007/s11356-022-21961-w] [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: 03/26/2022] [Accepted: 07/07/2022] [Indexed: 06/15/2023]
Abstract
Gestational arsenic (As) exposure is associated with intrauterine growth restriction (IUGR). This study explored the association among gestational As exposure, IUGR, and reduction of folate content in maternal and umbilical plasma from 530 mother-and-singleton-offspring pairs. Birth weight (BW) was negatively correlated with As in maternal plasma (r=-0.194, P<0.001) and umbilical plasma (r=-0.235, P<0.001). By contrast, a positive correlation was found between BW and maternal folate content (r=0.198, P<0.001). The subjects were divided into As-L and As-H groups. The influence of As-H on small for gestational age (SGA) infants, a marker of IUGR, was evaluated by multivariate logistic regression that excludes interferences of gestational age, infant sex, and other confounding factors. Mothers with As-H had an elevated risk of SGA infants (adjusted OR, 2.370; P<0.05). Interestingly, maternal folate content was lower in subjects with As-H than those with As-L (22.4±10.7 vs 11.2±6.7 nmol/L, P<0.001). Linear correlation models show that As level was negatively correlated with folate content in maternal plasma (r=-0.615, P<0.001) and umbilical plasma (r=-0.209, P<0.001). Moreover, maternal folate reduction has an obvious mediating effect between increased As and decreased BW (β=-0.078, P<0.05). Our results indicate that folate reduction may be a mediator between gestational As exposure and IUGR.
Collapse
Affiliation(s)
- Fei-Xiang Xu
- Department of Toxicology, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Xu Chen
- Department of Toxicology, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Heng Zhang
- Department of Toxicology, Anhui Medical University, Hefei, 230032, Anhui Province, China
- Wuxi Maternity and Child Health Care Hospital, Wuxi, 214122, Jiangsu Province, China
| | - Yi-Jun Fan
- Second Affiliated Hospital, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Ya-Ping Song
- Department of Toxicology, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Jin-Wei Lv
- Department of Toxicology, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Ya-Li Xie
- Department of Nutrition, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Yichao Huang
- Department of Toxicology, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Dao-Zhen Chen
- Wuxi Maternity and Child Health Care Hospital, Wuxi, 214122, Jiangsu Province, China
| | - Hua Wang
- Department of Toxicology, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - De-Xiang Xu
- Department of Toxicology, Anhui Medical University, Hefei, 230032, Anhui Province, China.
| |
Collapse
|
3
|
Caiaffa CD, Fonteles CSR, Yunping L, Finnell RH. Gene-environment interactions underlying the etiology of neural tube defects. Curr Top Dev Biol 2022; 152:193-220. [PMID: 36707212 DOI: 10.1016/bs.ctdb.2022.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Neural tube defects (NTDs) consist of severe structural malformations of the brain and spinal cord and are the second most common structural birth defect in humans, accounting for approximately 2700 affected pregnancies every year in the United States. These numbers are highly significant, considering that birth defects remain a leading cause of infant mortality in the United States, affecting approximately 120,000 babies born annually. Survivors of these congenital malformations face long-term disability and lifelong challenges imposed by severe physical burdens compromising the afflicted individual's overall quality of life. Clearly, birth defects, and especially NTDs remain a global public health challenge, and the source of significant financial repercussions for healthcare systems worldwide. In order to better understand the role gene-environment interactions play in the etiology of NTDs, this chapter provides an overview of NTD phenotypes and their embryonic origins, discusses the genetic landscape of NTDs as it is currently understood, with a focus on experimental models that best illustrate how environmental factors modulate individual susceptibility to these birth defects. As folic acid interventions have proven to be effective in reducing the prevalence of NTDs, the chapter ends with a discussion on the impact that maternal dietary status has on NTD prevalence from a population perspective.
Collapse
Affiliation(s)
- Carlo Donato Caiaffa
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Cristiane Sá Roriz Fonteles
- Postgraduate Program in Dentistry, Faculty of Pharmacy, Dentistry and Nursing, Federal University of Ceara, Fortaleza, Brazil
| | - Lei Yunping
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Richard H Finnell
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States; Departments of Molecular and Cellular Biology, Molecular and Human Genetics, and Medicine, Baylor College of Medicine, Houston, TX, United States.
| |
Collapse
|
4
|
Lv JW, Song YP, Zhang ZC, Fan YJ, Xu FX, Gao L, Zhang XY, Zhang C, Wang H, Xu DX. Gestational arsenic exposure induces anxiety-like behaviors in adult offspring by reducing DNA hydroxymethylation in the developing brain. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 227:112901. [PMID: 34673408 DOI: 10.1016/j.ecoenv.2021.112901] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/28/2021] [Accepted: 10/11/2021] [Indexed: 06/13/2023]
Abstract
Several studies found that reduction of 5-hydroxymethylcytosine (5hmC), a marker of DNA hydroxymethylation highly enriched in developing brain, is associated with anxiety-like behaviors. This study aimed to investigate whether gestational arsenic (As) exposure induces anxiety-like behaviors in adult offspring by reducing DNA hydroxymethylation in the developing brain. The dams drank ultrapure water containing NaAsO2 (15 mg/L) throughout pregnancy. Anxiety-like behaviors were evaluated and developing brain 5hmC was detected. Results showed that anxiety-like behaviors were observed in As-exposed adult offspring. In addition, 5hmC content was reduced in As-exposed fetal brain. Despite no difference on Tet1, Tet2 and Tet3 expression, TET activity was suppressed in As-exposed fetal brain. Mechanistically, alpha-ketoglutarate (α-KG), a cofactor for TET dioxygenases, was reduced and Idh2, a key enzymatic gene for mitochondrial α-KG synthesis, was downregulated in As-exposed fetal brain. Of interest, ascorbic acid, a cofactor for TET dioxygenases, reversed As-induced suppression of TET activity. Moreover, ascorbic acid attenuated As-induced reduction of 5hmC in fetal brain. In addition, ascorbic acid alleviated As-induced anxiety-like behaviors in adult offspring. Taken together, these results suggest that gestational As exposure induces anxiety-like behaviors in adult offspring, possibly at part, by inhibiting DNA hydroxymethylation in developing brain.
Collapse
Affiliation(s)
- Jin-Wei Lv
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Ya-Ping Song
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Zhi-Cheng Zhang
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Yi-Jun Fan
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Fei-Xiang Xu
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Lan Gao
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Xiao-Yi Zhang
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Cheng Zhang
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Hua Wang
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - De-Xiang Xu
- Department of Toxicology, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
5
|
Bae S, Kamynina E, Guetterman HM, Farinola AF, Caudill MA, Berry RJ, Cassano PA, Stover PJ. Provision of folic acid for reducing arsenic toxicity in arsenic-exposed children and adults. Cochrane Database Syst Rev 2021; 10:CD012649. [PMID: 34661903 PMCID: PMC8522704 DOI: 10.1002/14651858.cd012649.pub2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Arsenic is a common environmental toxin. Exposure to arsenic (particularly its inorganic form) through contaminated food and drinking water is an important public health burden worldwide, and is associated with increased risk of neurotoxicity, congenital anomalies, cancer, and adverse neurodevelopment in children. Arsenic is excreted following methylation reactions, which are mediated by folate. Provision of folate through folic acid supplements could facilitate arsenic methylation and excretion, thereby reducing arsenic toxicity. OBJECTIVES To assess the effects of provision of folic acid (through fortified foods or supplements), alone or in combination with other nutrients, in lessening the burden of arsenic-related health outcomes and reducing arsenic toxicity in arsenic-exposed populations. SEARCH METHODS In September 2020, we searched CENTRAL, MEDLINE, Embase, 10 other international databases, nine regional databases, and two trials registers. SELECTION CRITERIA Randomised controlled trials (RCTs) and quasi-RCTs comparing the provision of folic acid (at any dose or duration), alone or in combination with other nutrients or nutrient supplements, with no intervention, placebo, unfortified food, or the same nutrient or supplements without folic acid, in arsenic-exposed populations of all ages and genders. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. MAIN RESULTS We included two RCTs with 822 adults exposed to arsenic-contaminated drinking water in Bangladesh. The RCTs compared 400 µg/d (FA400) or 800 µg/d (FA800) folic acid supplements, given for 12 or 24 weeks, with placebo. One RCT, a multi-armed trial, compared FA400 plus creatine (3 g/d) to creatine alone. We judged both RCTs at low risk of bias in all domains. Due to differences in co-intervention, arsenic exposure, and participants' nutritional status, we could not conduct meta-analyses, and therefore, provide a narrative description of the data. Neither RCT reported on cancer, all-cause mortality, neurocognitive function, or congenital anomalies. Folic acid supplements alone versus placebo Blood arsenic. In arsenic-exposed individuals, FA likely reduces blood arsenic concentrations compared to placebo (2 studies, 536 participants; moderate-certainty evidence). For folate-deficient and folate-replete participants who received arsenic-removal water filters as a co-intervention, FA800 reduced blood arsenic levels more than placebo (percentage change (%change) in geometric mean (GM) FA800 -17.8%, 95% confidence intervals (CI) -25.0 to -9.8; placebo GM -9.5%, 95% CI -16.5 to -1.8; 1 study, 406 participants). In one study with 130 participants with low baseline plasma folate, FA400 reduced total blood arsenic (%change FA400 mean (M) -13.62%, standard error (SE) ± 2.87; placebo M -2.49%, SE ± 3.25), and monomethylarsonic acid (MMA) concentrations (%change FA400 M -22.24%, SE ± 2.86; placebo M -1.24%, SE ± 3.59) more than placebo. Inorganic arsenic (InAs) concentrations reduced in both groups (%change FA400 M -18.54%, SE ± 3.60; placebo M -10.61%, SE ± 3.38). There was little to no change in dimethylarsinic acid (DMA) in either group. Urinary arsenic. In arsenic-exposed individuals, FA likely reduces the proportion of total urinary arsenic excreted as InAs (%InAs) and MMA (%MMA) and increases the proportion excreted as DMA (%DMA) to a greater extent than placebo (2 studies, 546 participants; moderate-certainty evidence), suggesting that FA enhances arsenic methylation. In a mixed folate-deficient and folate-replete population (1 study, 352 participants) receiving arsenic-removal water filters as a co-intervention, groups receiving FA had a greater decrease in %InAs (within-person change FA400 M -0.09%, 95% CI -0.17 to -0.01; FA800 M -0.14%, 95% CI -0.21 to -0.06; placebo M 0.05%, 95% CI 0.00 to 0.10), a greater decrease in %MMA (within-person change FA400 M -1.80%, 95% CI -2.53 to -1.07; FA800 M -2.60%, 95% CI -3.35 to -1.85; placebo M 0.15%, 95% CI -0.37 to 0.68), and a greater increase in %DMA (within-person change FA400 M 3.25%, 95% CI 1.81 to 4.68; FA800 M 4.57%, 95% CI 3.20 to 5.95; placebo M -1.17%, 95% CI -2.18 to -0.17), compared to placebo. In 194 participants with low baseline plasma folate, FA reduced %InAs (%change FA400 M -0.31%, SE ± 0.04; placebo M -0.13%, SE ± 0.04) and %MMA (%change FA400 M -2.6%, SE ± 0.37; placebo M -0.71%, SE ± 0.43), and increased %DMA (%change FA400 M 5.9%, SE ± 0.82; placebo M 2.14%, SE ± 0.71), more than placebo. Plasma homocysteine: In arsenic-exposed individuals, FA400 likely reduces homocysteine concentrations to a greater extent than placebo (2 studies, 448 participants; moderate-certainty evidence), in the mixed folate-deficient and folate-replete population receiving arsenic-removal water filters as a co-intervention (%change in GM FA400 -23.4%, 95% CI -27.1 to -19.5; placebo -1.3%, 95% CI -5.3 to 3.1; 1 study, 254 participants), and participants with low baseline plasma folate (within-person change FA400 M -3.06 µmol/L, SE ± 3.51; placebo M -0.05 µmol/L, SE ± 4.31; 1 study, 194 participants). FA supplements plus other nutrient supplements versus nutrient supplements alone In arsenic-exposed individuals who received arsenic-removal water filters as a co-intervention, FA400 plus creatine may reduce blood arsenic concentrations more than creatine alone (%change in GM FA400 + creatine -14%, 95% CI -22.2 to -5.0; creatine -7.0%, 95% CI -14.8 to 1.5; 1 study, 204 participants; low-certainty evidence); may not change urinary arsenic methylation indices (FA400 + creatine: %InAs M 13.2%, SE ± 7.0; %MMA M 10.8, SE ± 4.1; %DMA M 76, SE ± 7.8; creatine: %InAs M 14.8, SE ± 5.5; %MMA M 12.8, SE ± 4.0; %DMA M 72.4, SE ±7.6; 1 study, 190 participants; low-certainty evidence); and may reduce homocysteine concentrations to a greater extent (%change in GM FA400 + creatinine -21%, 95% CI -25.2 to -16.4; creatine -4.3%, 95% CI -9.0 to 0.7; 1 study, 204 participants; low-certainty evidence) than creatine alone. AUTHORS' CONCLUSIONS There is moderate-certainty evidence that FA supplements may benefit blood arsenic concentration, urinary arsenic methylation profiles, and plasma homocysteine concentration versus placebo. There is low-certainty evidence that FA supplements plus other nutrients may benefit blood arsenic and plasma homocysteine concentrations versus nutrients alone. No studies reported on cancer, all-cause mortality, neurocognitive function, or congenital anomalies. Given the limited number of RCTs, more studies conducted in diverse settings are needed to assess the effects of FA on arsenic-related health outcomes and arsenic toxicity in arsenic-exposed adults and children.
Collapse
Affiliation(s)
- Sajin Bae
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Elena Kamynina
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | | | - Adetutu F Farinola
- Faculty of Public Health, Department of Human Nutrition and Dietetics, University of Ibadan, Ibadan, Nigeria
| | - Marie A Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Robert J Berry
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | |
Collapse
|
6
|
Resveratrol attenuates arsenic-induced cognitive deficits via modulation of Estrogen-NMDAR-BDNF signalling pathway in female mouse hippocampus. Psychopharmacology (Berl) 2021; 238:2485-2502. [PMID: 34050381 DOI: 10.1007/s00213-021-05871-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/05/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Chronic inorganic arsenic (iAs) exposure induces deleterious effects on CNS including oxidative stress, cognitive deficits and altered brain neurochemistry. Little is known about the association between iAs and estrogen receptor expression in brain regions. AIMS AND OBJECTIVES Owing to the neuroprotective and estrogenic activities of resveratrol (RES), we examined the combined effects of arsenic trioxide (As2O3) and RES on neurobehavioural functions, estrogen signalling and associated neurochemical changes in mouse hippocampus. MATERIALS AND METHODS As2O3 alone (2 and 4 mg/kg bw) or along with RES (40 mg/kg bw) was administered orally for 45 days to adult female mice. From days 33 to 45, open field, elevated plus maze and Morris water maze tests were conducted to evaluate locomotion, anxiety and learning and memory. On day 46, animals were euthanized and brain tissue and hippocampi obtained therefrom were processed for atomic absorption spectrophotometry and western blotting respectively. RESULTS As2O3 alone exposure resulted in enhanced anxiety levels, reduced locomotion and impaired learning and memory. As2O3-induced behavioural deficits were accompanied by downregulation of estrogen receptor (ERα) expression with a concomitant reduction of BDNF and NMDAR 2B levels in the hippocampus. However, the behavioural alterations and expression of these markers were restored in RES-supplemented mice. Moreover, a dose-dependent iAs accumulation was observed in serum and brain tissues of mice receiving As2O3 alone whereas simultaneous administration of As2O3 with RES facilitated iAs efflux. CONCLUSIONS These results suggest that reduced ERα expression with associated downregulation of BDNF and NMDAR 2B levels could be a mechanism by which iAs induces cognitive impairment; hence, the modulation of estrogen-NMDAR-BDNF pathway by RES represents a potential avenue to recover behavioural deficits induced by this neurotoxin.
Collapse
|
7
|
Chen WJ, Davis EM, Stoner JA, Robledo C, Goodman JR, Garwe T, Janitz AE, Xu C, Hwang J, Peck JD. Urinary total arsenic and arsenic methylation capacity in pregnancy and gestational diabetes mellitus: A case-control study. CHEMOSPHERE 2021; 271:129828. [PMID: 33736216 PMCID: PMC8966639 DOI: 10.1016/j.chemosphere.2021.129828] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 05/12/2023]
Abstract
Previous studies suggest arsenic exposure may increase the risk of gestational diabetes mellitus (GDM). However, prior assessments of total arsenic concentrations have not distinguished between toxic and nontoxic species. Our study aimed to investigate the relationships between inorganic arsenic exposure, arsenic methylation capacity, and GDM. Sixty-four cases of GDM and 237 controls were analyzed for urinary concentrations of inorganic arsenic species and their metabolites (arsenite (As3), arsenate (As5), monomethylarsonic acid (MMA), and dimethylarsinic acid (DMA)), and organic forms of arsenic. Inorganic arsenic exposure was defined as the sum of inorganic and methylated arsenic species (iSumAs). Methylation capacity indices were calculated as the percentage of inorganic arsenic species [iAs% = (As3 + As5)/iSumAs, MMA% = MMA/iSumAs, and DMA% = DMA/iSumAs]. Multivariable logistic regression was performed to evaluate the association between inorganic arsenic exposure, methylation capacity indices, and GDM. We did not observe evidence of a positive association between iSumAs and GDM. However, women with GDM had an increased odds of inefficient methylation capacity when comparing the highest and lowest tertiles of iAs% (adjusted odds ratio (aOR) = 1.48, 95% CI 0.58-3.77) and MMA% (aOR = 1.95 (95% CI 0.81-4.70) and a reduced odds of efficient methylation capacity as indicated by DMA% (aOR = 0.62 (95% CI 0.25-1.52), though the confidence intervals included the null value. While the observed associations with arsenic methylation indices were imprecise and warrant cautious interpretation, the direction and magnitude of the relative measures reflected a pattern of lower detoxification of inorganic arsenic exposures among women with GDM.
Collapse
Affiliation(s)
- Wei-Jen Chen
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Erin M Davis
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, KY, USA
| | - Julie A Stoner
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Candace Robledo
- Department of Population Health and Biostatistics, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Jean R Goodman
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Loyola University Medical Center, Maywood, IL, USA
| | - Tabitha Garwe
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Amanda E Janitz
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Chao Xu
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jooyeon Hwang
- Department of Occupational and Environmental Health, Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jennifer D Peck
- Department of Biostatistics and Epidemiology, Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
8
|
Varshavsky J, Smith A, Wang A, Hom E, Izano M, Huang H, Padula A, Woodruff TJ. Heightened susceptibility: A review of how pregnancy and chemical exposures influence maternal health. Reprod Toxicol 2020; 92:14-56. [PMID: 31055053 PMCID: PMC6824944 DOI: 10.1016/j.reprotox.2019.04.004] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 03/12/2019] [Accepted: 04/22/2019] [Indexed: 12/19/2022]
Abstract
Pregnancy is a unique period when biological changes can increase sensitivity to chemical exposures. Pregnant women are exposed to multiple environmental chemicals via air, food, water, and consumer products, including flame retardants, plasticizers, and pesticides. Lead exposure increases risk of pregnancy-induced hypertensive disorders, although women's health risks are poorly characterized for most chemicals. Research on prenatal exposures has focused on fetal outcomes and less on maternal outcomes. We reviewed epidemiologic literature on chemical exposures during pregnancy and three maternal outcomes: preeclampsia, gestational diabetes, and breast cancer. We found that pregnancy can heighten susceptibility to environmental chemicals and women's health risks, although variations in study design and exposure assessment limited study comparability. Future research should include pregnancy as a critical period for women's health. Incorporating biomarkers of exposure and effect, deliberate timing and method of measurement, and consistent adjustment of potential confounders would strengthen research on the exposome and women's health.
Collapse
Affiliation(s)
- Julia Varshavsky
- University of California, San Francisco, Program on Reproductive Health and the Environment, San Francisco, CA, USA.
| | - Anna Smith
- University of California, Berkeley, School of Public Health, Berkeley, CA, USA
| | - Aolin Wang
- University of California, San Francisco, Program on Reproductive Health and the Environment, San Francisco, CA, USA; University of California, San Francisco, Bakar Computational Health Sciences Institute, San Francisco, CA, USA
| | - Elizabeth Hom
- University of California, San Francisco, Program on Reproductive Health and the Environment, San Francisco, CA, USA
| | - Monika Izano
- University of California, San Francisco, Program on Reproductive Health and the Environment, San Francisco, CA, USA
| | - Hongtai Huang
- University of California, San Francisco, Program on Reproductive Health and the Environment, San Francisco, CA, USA; University of California, San Francisco, Bakar Computational Health Sciences Institute, San Francisco, CA, USA
| | - Amy Padula
- University of California, San Francisco, Program on Reproductive Health and the Environment, San Francisco, CA, USA
| | - Tracey J Woodruff
- University of California, San Francisco, Program on Reproductive Health and the Environment, San Francisco, CA, USA
| |
Collapse
|
9
|
Sargis RM, Simmons RA. Environmental neglect: endocrine disruptors as underappreciated but potentially modifiable diabetes risk factors. Diabetologia 2019; 62:1811-1822. [PMID: 31451869 PMCID: PMC7462102 DOI: 10.1007/s00125-019-4940-z] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 05/14/2019] [Indexed: 12/18/2022]
Abstract
Type 2 diabetes prevalence is increasing dramatically across the globe, imposing a tremendous toll on individuals and healthcare systems. Reversing these trends requires comprehensive approaches to address both classical and emerging diabetes risk factors. Recently, environmental toxicants acting as endocrine-disrupting chemicals (EDCs) have emerged as novel metabolic disease risk factors. EDCs implicated in diabetes pathogenesis include various inorganic and organic molecules of both natural and synthetic origin, including arsenic, bisphenol A, phthalates, polychlorinated biphenyls and organochlorine pesticides. Indeed, evidence implicates EDC exposures across the lifespan in metabolic dysfunction; moreover, specific developmental windows exhibit enhanced sensitivity to EDC-induced metabolic disruption, with potential impacts across generations. Importantly, differential exposures to diabetogenic EDCs likely also contribute to racial/ethnic and economic disparities. Despite these emerging links, clinical practice guidelines fail to address this underappreciated diabetes risk factor. Comprehensive approaches to stem the tide of diabetes must include efforts to address its environmental drivers.
Collapse
Affiliation(s)
- Robert M Sargis
- Division of Endocrinology, Diabetes, and Metabolism Department of Medicine, University of Illinois at Chicago, 835 S. Wolcott, Suite E625; M/C 640, Chicago, IL, 60612, USA.
- ChicAgo Center for Health and EnvironmenT (CACHET), University of Illinois at Chicago, Chicago, IL, USA.
| | - Rebecca A Simmons
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Research on Reproduction and Women's Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
10
|
Gong Y, Liu J, Xue Y, Zhuang Z, Qian S, Zhou W, Li X, Qian J, Ding G, Sun Z. Non-monotonic dose-response effects of arsenic on glucose metabolism. Toxicol Appl Pharmacol 2019; 377:114605. [PMID: 31170414 DOI: 10.1016/j.taap.2019.114605] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/29/2019] [Accepted: 05/31/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Inorganic arsenic (iAs) is a widespread environmental toxin. In addition to being a human carcinogen, its effect on diabetes has started to gain recognition recently. Insulin is the key hormone regulating systemic glucose metabolism. The in vivo effect of iAs on insulin sensitivity has not been directly addressed. OBJECTIVES Here we use mouse models to dissect the dose-dependent effects of iAs on glucose metabolism in vivo. METHODS We performed hyperinsulinemic-euglycemic clamp, the gold standard analysis of systemic insulin sensitivity. We also performed dynamic metabolic testings and RNA-seq analysis. RESULTS We found that a low-dose exposure (0.25 ppm iAs in drinking water) caused glucose intolerance in adult male C57BL/6 mice, likely by disrupting glucose-induced insulin secretion without affecting peripheral insulin sensitivity. However, a higher-dose exposure (2.5 ppm iAs) had diminished effects on glucose tolerance despite disrupted pancreatic insulin secretion. Insulin Clamp analysis showed that 2.5 ppm iAs actually enhanced systemic insulin sensitivity by simultaneously enhancing insulin-stimulated glucose uptake in skeletal muscles and improved insulin-mediated suppression of endogenous glucose production. RNA-seq analysis of skeletal muscles revealed that 2.5 ppm iAs regulated expression of many genes involved in the metabolism of fatty acids, pyruvate, and amino acids. CONCLUSION These findings suggest that iAs has opposite glycemic effects on distinct metabolic tissues at different dose thresholds. Such non-monotonic dose-response effects of iAs on glucose tolerance shed light on the complex interactions between iAs and the systemic glucose metabolism, which could potentially help reconcile some of the conflicting results in human epidemiological studies.
Collapse
Affiliation(s)
- Yingyun Gong
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Jidong Liu
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Yanfeng Xue
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Zhong Zhuang
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Sichong Qian
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Wenjun Zhou
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Xin Li
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Justin Qian
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Guolian Ding
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America; The International Peace Maternity and Child Health Hospital, Institute of Embryo-Fetal Original Adult Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zheng Sun
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America.
| |
Collapse
|
11
|
Potential facet for prenatal arsenic exposure paradigm: linking endocrine disruption and epigenetics. THE NUCLEUS 2019. [DOI: 10.1007/s13237-019-00274-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
12
|
Carmean CM, Seino S. Braving the Element: Pancreatic β-Cell Dysfunction and Adaptation in Response to Arsenic Exposure. Front Endocrinol (Lausanne) 2019; 10:344. [PMID: 31258514 PMCID: PMC6587364 DOI: 10.3389/fendo.2019.00344] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 05/13/2019] [Indexed: 12/26/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a serious global health problem, currently affecting an estimated 451 million people worldwide. T2DM is characterized by hyperglycemia and low insulin relative to the metabolic demand. The precise contributing factors for a given individual vary, but generally include a combination of insulin resistance and insufficient insulin secretion. Ultimately, the progression to diabetes occurs only after β-cells fail to meet the needs of the individual. The stresses placed upon β-cells in this context manifest as increased oxidative damage, local inflammation, and ER stress, often inciting a destructive spiral of β-cell death, increased metabolic stress due to further insufficiency, and additional β-cell death. Several pathways controlling insulin resistance and β-cell adaptation/survival are affected by a class of exogenous bioactive compounds deemed endocrine disrupting chemicals (EDCs). Epidemiological studies have shown that, in several regions throughout the world, exposure to the EDC inorganic arsenic (iAs) correlates significantly with T2DM. It has been proposed that a lifetime of exposure to iAs may exacerbate problems with both insulin sensitivity as well as β-cell function/survival, promoting the development of T2DM. This review focuses on the mechanisms of iAs action as they relate to known adaptive and maladaptive pathways in pancreatic β-cells.
Collapse
Affiliation(s)
- Christopher M. Carmean
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- *Correspondence: Christopher M. Carmean
| | - Susumu Seino
- Division of Molecular and Metabolic Medicine, Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Kobe, Japan
- Susumu Seino
| |
Collapse
|
13
|
Mazumdar M. Does arsenic increase the risk of neural tube defects among a highly exposed population? A new case-control study in Bangladesh. Birth Defects Res 2018; 109:92-98. [PMID: 27801974 DOI: 10.1002/bdra.23577] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 08/01/2016] [Accepted: 08/12/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND Neural tube defects are debilitating birth defects that occur when the developing neural plate fails to close in early gestation. Arsenic induces neural tube defects in animal models, but whether environmental arsenic exposure increases risk of neural tube defects in humans is unknown. METHODS We describe a new case-control study in Bangladesh, a country currently experiencing an epidemic of arsenic poisoning through contaminated drinking water. We plan to understand how arsenic influences risk of neural tube defects in humans through mechanisms that include disruption of maternal glucose and folate metabolism, as well as epigenetic effects. We also investigate whether sweat chloride concentration, a potential new biomarker for arsenic toxicity, can be used to identify women at higher risk for having a child affected by neural tube defect. We will collect dural tissue from cases, obtained at the time of surgical closure of the defect, and believe investigation of these samples will provide insight into the epigenetic mechanisms by which prenatal arsenic exposure affects the developing nervous system. CONCLUSION These studies explore mechanisms by which arsenic may increase risk of neural tube defects in humans and use a unique population with high arsenic exposure to test hypotheses. If successful, these studies may assist countries with high arsenic exposure such as Bangladesh to identify populations at high risk of neural tube defects, as well as direct development of novel screening strategies for maternal risk.Birth Defects Research 109:92-98, 2017.© 2016 The Authors Birth Defects Research Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Maitreyi Mazumdar
- Department of Neurology, Boston Children's Hospital, Boston, Massachusetts.,Department of Neurology, Harvard Medical School, Boston, Massachusetts.,Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
14
|
Demir N, Başaranoğlu M, Huyut Z, Değer İ, Karaman K, Şekeroğlu MR, Tuncer O. The relationship between mother and infant plasma trace element and heavy metal levels and the risk of neural tube defect in infants. J Matern Fetal Neonatal Med 2017; 32:1433-1440. [DOI: 10.1080/14767058.2017.1408064] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Nihat Demir
- Department of Pediatrics, Division of Neonatology, Yuzuncu Yil University School of Medicine, Van, Turkey
| | - Murat Başaranoğlu
- Department of Pediatrics, Division of Neonatology, Yuzuncu Yil University School of Medicine, Van, Turkey
| | - Zübeyir Huyut
- Department of Medical Biochemistry, Yuzuncu Yil University School of Medicine, Van, Turkey
| | - İbrahim Değer
- Department of Pediatrics, Division of Neonatology, Yuzuncu Yil University School of Medicine, Van, Turkey
| | - Kamuran Karaman
- Department of Pediatrics, Division of Neonatology, Yuzuncu Yil University School of Medicine, Van, Turkey
| | - M. Ramazan Şekeroğlu
- Department of Medical Biochemistry, Yuzuncu Yil University School of Medicine, Van, Turkey
| | - Oğuz Tuncer
- Department of Pediatrics, Division of Neonatology, Yuzuncu Yil University School of Medicine, Van, Turkey
| |
Collapse
|
15
|
Polluted Pathways: Mechanisms of Metabolic Disruption by Endocrine Disrupting Chemicals. Curr Environ Health Rep 2017; 4:208-222. [PMID: 28432637 DOI: 10.1007/s40572-017-0137-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Environmental toxicants are increasingly implicated in the global decline in metabolic health. Focusing on diabetes, herein, the molecular and cellular mechanisms by which metabolism disrupting chemicals (MDCs) impair energy homeostasis are discussed. RECENT FINDINGS Emerging data implicate MDC perturbations in a variety of pathways as contributors to metabolic disease pathogenesis, with effects in diverse tissues regulating fuel utilization. Potentiation of traditional metabolic risk factors, such as caloric excess, and emerging threats to metabolism, such as disruptions in circadian rhythms, are important areas of current and future MDC research. Increasing evidence also implicates deleterious effects of MDCs on metabolic programming that occur during vulnerable developmental windows, such as in utero and early post-natal life as well as pregnancy. Recent insights into the mechanisms by which MDCs alter energy homeostasis will advance the field's ability to predict interactions with classical metabolic disease risk factors and empower studies utilizing targeted therapeutics to treat MDC-mediated diabetes.
Collapse
|
16
|
Bonini MG, Sargis RM. Environmental Toxicant Exposures and Type 2 Diabetes Mellitus: Two Interrelated Public Health Problems on the Rise. CURRENT OPINION IN TOXICOLOGY 2017; 7:52-59. [PMID: 29392186 DOI: 10.1016/j.cotox.2017.09.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Rates of type 2 diabetes mellitus (T2DM) are rising rapidly across the globe and the impact of this devastating disease threatens to plague the 21st century. While some contributing factors are well-recognized (e.g. sedentary lifestyles and caloric excess), others diabetes-promoting risk factors are less established or poorly appreciated. The latter category includes environmental exposures to diabetogenic contaminants. Herein we review some of the latest concepts and mechanisms by which environmental exposures may contribute to rising rates of T2DM with a particular focus on mechanisms involving mitochondrial dysfunction and imbalances in reactive oxygen species (ROS). Furthermore, while the pathogenesis of diabetes includes impairments in insulin sensitivity as well as insulin secretion, we will specifically delve into the links between environmental exposures to toxicants such as arsenic and disruptions in insulin release from pancreatic β-cells. Since β-cell death or dysfunction lies at the heart of both T2DM as well as type 1 diabetes mellitus (T1DM), environmental endocrine disrupting chemicals (EDCs) that disrupt the production or regulated release of the glucose-lowering hormone insulin are likely contributors to diabetes risk. Importantly, understanding the contribution of toxicants to diabetes risk as well as improved understanding of their mechanisms of action offer unique opportunities to modulate diabetes risk via targeted therapeutics or public policy interventions to reduce and remediate exposures.
Collapse
Affiliation(s)
- Marcelo G Bonini
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Robert M Sargis
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
17
|
Jahanbazi Jahan-Abad A, Morteza-zadeh P, Sahab Negah S, Gorji A. Curcumin attenuates harmful effects of arsenic on neural stem/progenitor cells. AVICENNA JOURNAL OF PHYTOMEDICINE 2017; 7:376-388. [PMID: 28884087 PMCID: PMC5580875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/08/2016] [Accepted: 12/24/2016] [Indexed: 11/02/2022]
Abstract
OBJECTIVE Arsenic, an environmental pollutant, decreases neuronal migration as well as cellular maturation and inhibits the proliferation of neural progenitor cells. Curcumin has been described as an antioxidant and neuroprotective agent with strong therapeutic potential in some neurological disorders. Human adipose-derived stem cells (hADSCs), a source of multipotent stem cells, can self-renew and differentiate into neural cells. The aim of the present study was to investigate the preventive effect of curcumin against arsenic toxic effects on the viability, telomerase activity, and apoptosis of neural stem/progenitor cells (NSPCs) derived from hADSCs. MATERIALS AND METHODS The characteristics of human adipose tissue were identified by immunocytochemistry for surface markers namely, CD105, CD73, and CD90. Using neurosphere assay, hADSCs were differentiated into neuronal cells. To characterize neural cells, expression of nestin, SOX2, MAP2, and GFAP were assessed by immunocytochemistry. Cytotoxicity and viability of NSPCs were evaluated by MTT assay. Reactive oxygen species (ROS) generated by arsenic exposure, were measured and caspase 3/7 activity and caspase-3 processing as well as the telomerase activity were determined. RESULTS The isolated hADSCs positively expressed CD105, CD73, and CD90. Nestin, Sox2, GFAP, and MAP2 were expressed in the neurospheres derived from hADSCs. Curcumin/arsenic co-treatment significantly increased telomerase activity of NSPCs compared to arsenic group. Furthermore, curcumin significantly reduced arsenic-induced apoptosis (via inactivation of caspases) as well as arsenic-associated ROS generation. CONCLUSION Our findings revealed that curcumin has the potential to prevent harmful effects of arsenic on neurogenesis.
Collapse
Affiliation(s)
- Ali Jahanbazi Jahan-Abad
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
- Department of Clinical Biochemistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Sajad Sahab Negah
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
- Department of Neuroscience, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
- Department of Neuroscience, Mashhad University of Medical Sciences, Mashhad, Iran
- Epilepsy Research Center, Department of Neurology, and Department of Neurosurgery, Westfälische Wilhelms-Universität Münster, Münster, Germany
| |
Collapse
|
18
|
Bae S, Kamynina E, Farinola AF, Caudill MA, Stover PJ, Cassano PA, Berry R, Peña-Rosas JP. Provision of folic acid for reducing arsenic toxicity in arsenic-exposed children and adults. THE COCHRANE DATABASE OF SYSTEMATIC REVIEWS 2017. [DOI: 10.1002/14651858.cd012649] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Sajin Bae
- Cornell University; Division of Nutritional Sciences; 324 Savage Hall 244 Garden Avenue Ithaca NY USA 14853
| | - Elena Kamynina
- Cornell University; Division of Nutritional Sciences; 324 Savage Hall 244 Garden Avenue Ithaca NY USA 14853
| | - Adetutu F Farinola
- University of Ibadan; Faculty of Public Health, Department of Human Nutrition and Dietetics; Seat of Wisdom Chapel Ibadan Oyo State Nigeria 200282
| | - Marie A Caudill
- Cornell University; Division of Nutritional Sciences; 324 Savage Hall 244 Garden Avenue Ithaca NY USA 14853
| | - Patrick J Stover
- Cornell University; Division of Nutritional Sciences; 324 Savage Hall 244 Garden Avenue Ithaca NY USA 14853
| | - Patricia A Cassano
- Cornell University; Division of Nutritional Sciences; 324 Savage Hall 244 Garden Avenue Ithaca NY USA 14853
| | - Robert Berry
- Independent cosultant; 1376 N Decatur Rd NE Atlanta Georgia USA 30306
| | - Juan Pablo Peña-Rosas
- World Health Organization; Evidence and Programme Guidance, Department of Nutrition for Health and Development; 20 Avenue Appia Geneva GE Switzerland 1211
| |
Collapse
|
19
|
Heindel JJ, Blumberg B, Cave M, Machtinger R, Mantovani A, Mendez MA, Nadal A, Palanza P, Panzica G, Sargis R, Vandenberg LN, Vom Saal F. Metabolism disrupting chemicals and metabolic disorders. Reprod Toxicol 2017; 68:3-33. [PMID: 27760374 PMCID: PMC5365353 DOI: 10.1016/j.reprotox.2016.10.001] [Citation(s) in RCA: 705] [Impact Index Per Article: 88.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 09/04/2016] [Accepted: 10/13/2016] [Indexed: 01/09/2023]
Abstract
The recent epidemics of metabolic diseases, obesity, type 2 diabetes(T2D), liver lipid disorders and metabolic syndrome have largely been attributed to genetic background and changes in diet, exercise and aging. However, there is now considerable evidence that other environmental factors may contribute to the rapid increase in the incidence of these metabolic diseases. This review will examine changes to the incidence of obesity, T2D and non-alcoholic fatty liver disease (NAFLD), the contribution of genetics to these disorders and describe the role of the endocrine system in these metabolic disorders. It will then specifically focus on the role of endocrine disrupting chemicals (EDCs) in the etiology of obesity, T2D and NAFLD while finally integrating the information on EDCs on multiple metabolic disorders that could lead to metabolic syndrome. We will specifically examine evidence linking EDC exposures during critical periods of development with metabolic diseases that manifest later in life and across generations.
Collapse
Affiliation(s)
- Jerrold J Heindel
- National Institute of Environmental Health Sciences, Division of Extramural Research and Training Research Triangle Park, NC, USA.
| | - Bruce Blumberg
- University of California, Department of Developmental and Cell Biology, Irvine CA, USA
| | - Mathew Cave
- University of Louisville, Division of Gastroenterology, Hepatology and Nutrition, Louisville KY, USA
| | | | | | - Michelle A Mendez
- University of North Carolina at Chapel Hill, School of Public Health, Chapel Hill NC, USA
| | - Angel Nadal
- Institute of Bioengineering and CIBERDEM, Miguel Hernandez University of Elche, Elche, Alicante, Spain
| | - Paola Palanza
- University of Parma, Department of Neurosciences, Parma, Italy
| | - Giancarlo Panzica
- University of Turin, Department of Neuroscience and Neuroscience Institute Cavalieri Ottolenghi (NICO), Turin, Italy
| | - Robert Sargis
- University of Chicago, Section of Endocrinology, Diabetes and Metabolism, Department of Medicine Chicago, IL, USA
| | - Laura N Vandenberg
- University of Massachusetts, Department of Environmental Health Sciences, School of Public Health & Health Sciences, Amherst, MA, USA
| | - Frederick Vom Saal
- University of Missouri, Department of Biological Sciences, Columbia, MO, USA
| |
Collapse
|
20
|
Rodriguez KF, Ungewitter EK, Crespo-Mejias Y, Liu C, Nicol B, Kissling GE, Yao HHC. Effects of in Utero Exposure to Arsenic during the Second Half of Gestation on Reproductive End Points and Metabolic Parameters in Female CD-1 Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:336-43. [PMID: 26295903 PMCID: PMC4786990 DOI: 10.1289/ehp.1509703] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 08/17/2015] [Indexed: 05/19/2023]
Abstract
BACKGROUND Mice exposed to high levels of arsenic in utero have increased susceptibility to tumors such as hepatic and pulmonary carcinomas when they reach adulthood. However, the effects of in utero arsenic exposure on general physiological functions such as reproduction and metabolism remain unclear. OBJECTIVES We evaluated the effects of in utero exposure to inorganic arsenic at the U.S. Environmental Protection Agency (EPA) drinking water standard (10 ppb) and at tumor-inducing levels (42.5 ppm) on reproductive end points and metabolic parameters when the exposed females reached adulthood. METHODS Pregnant CD-1 mice were exposed to sodium arsenite [none (control), 10 ppb, or 42.5 ppm] in drinking water from gestational day 10 to birth, the window of organ formation. At birth, exposed offspring were fostered to unexposed dams. We examined reproductive end points (age at vaginal opening, reproductive hormone levels, estrous cyclicity, and fertility) and metabolic parameters (body weight changes, hormone levels, body fat content, and glucose tolerance) in the exposed females when they reached adulthood. RESULTS Arsenic-exposed females (10 ppb and 42.5 ppm) exhibited early onset of vaginal opening. Fertility was not affected when females were exposed to the 10-ppb dose. However, the number of litters per female was decreased in females exposed to 42.5 ppm of arsenic in utero. In both 10-ppb and 42.5-ppm groups, arsenic-exposed females had significantly greater body weight gain, body fat content, and glucose intolerance. CONCLUSION Our findings revealed unexpected effects of in utero exposure to arsenic: exposure to both a human-relevant low dose and a tumor-inducing level led to early onset of vaginal opening and to obesity in female CD-1 mice.
Collapse
Affiliation(s)
- Karina F. Rodriguez
- Reproductive Developmental Biology Group, Reproductive and Developmental Biology Laboratory, and
| | - Erica K. Ungewitter
- Reproductive Developmental Biology Group, Reproductive and Developmental Biology Laboratory, and
| | - Yasmin Crespo-Mejias
- Reproductive Developmental Biology Group, Reproductive and Developmental Biology Laboratory, and
| | - Chang Liu
- Reproductive Developmental Biology Group, Reproductive and Developmental Biology Laboratory, and
| | - Barbara Nicol
- Reproductive Developmental Biology Group, Reproductive and Developmental Biology Laboratory, and
| | - Grace E. Kissling
- Biostatistics Branch, National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, USA
| | - Humphrey Hung-Chang Yao
- Reproductive Developmental Biology Group, Reproductive and Developmental Biology Laboratory, and
- Address correspondence to H.H.-C. Yao, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services, 111 T.W. Alexander Dr., C4-10, Research Triangle Park, NC 27709 USA, Telephone: (919) 541-1095. E-mail:
| |
Collapse
|
21
|
Robledo CA, Romano ME, Alonso-Magdalena P. Review of Current Evidence on the Impact of Environmental Chemicals on Gestational Diabetes Mellitus. CURR EPIDEMIOL REP 2016. [DOI: 10.1007/s40471-016-0070-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
22
|
Prenatal Exposure to Sodium Arsenite Alters Placental Glucose 1, 3, and 4 Transporters in Balb/c Mice. BIOMED RESEARCH INTERNATIONAL 2015; 2015:175025. [PMID: 26339590 PMCID: PMC4538324 DOI: 10.1155/2015/175025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 04/17/2015] [Indexed: 12/15/2022]
Abstract
Inorganic arsenic (iAs) exposure induces a decrease in glucose type 4 transporter (GLUT4) expression on the adipocyte membrane, which may be related to premature births and low birth weight infants in women exposed to iAs at reproductive age. The aim of this study was to analyze the effect of sodium arsenite (NaAsO2) exposure on GLUT1, GLUT3, and GLUT4 protein expression and on placental morphology. Female Balb/c mice (n = 15) were exposed to 0, 12, and 20 ppm of NaAsO2 in drinking water from 8th to 18th day of gestation. Morphological changes and GLUT1, GLUT3, and GLUT4 expression were evaluated in placentas by immunohistochemical and image analysis and correlated with iAs and arsenical species concentration, which were quantified by atomic absorption spectroscopy. NaAsO2 exposure induced a significant decrease in fetal and placental weight (P < 0.01) and increases in infarctions and vascular congestion. Whereas GLUT1 expression was unchanged in placentas from exposed group, GLUT3 expression was found increased. In contrast, GLUT4 expression was significantly lower (P < 0.05) in placentas from females exposed to 12 ppm. The decrease in placental GLUT4 expression might affect the provision of adequate fetal nutrition and explain the low fetal weight observed in the exposed groups.
Collapse
|
23
|
Mazumdar M, Ibne Hasan MOS, Hamid R, Valeri L, Paul L, Selhub J, Rodrigues EG, Silva F, Mia S, Mostofa MG, Quamruzzaman Q, Rahman M, Christiani DC. Arsenic is associated with reduced effect of folic acid in myelomeningocele prevention: a case control study in Bangladesh. Environ Health 2015; 14:34. [PMID: 25885259 PMCID: PMC4404044 DOI: 10.1186/s12940-015-0020-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 03/20/2015] [Indexed: 05/22/2023]
Abstract
BACKGROUND Arsenic induces neural tube defects in several animal models, but its potential to cause neural tube defects in humans is unknown. Our objective was to investigate the associations between maternal arsenic exposure, periconceptional folic acid supplementation, and risk of posterior neural tube defect (myelomeningocele) among a highly exposed population in rural Bangladesh. METHODS We performed a case-control study that recruited physician-confirmed cases from community health clinics served by Dhaka Community Hospital in Bangladesh, as well as local health facilities that treat children with myelomeningocele. Controls were selected from pregnancy registries in the same areas. Maternal arsenic exposure was estimated from drinking water samples taken from wells used during the first trimester of pregnancy. Periconceptional folic acid use was ascertained by self-report, and maternal folate status was further assessed by plasma folate levels measured at the time of the study visit. RESULTS Fifty-seven cases of myelomeningocele were identified along with 55 controls. A significant interaction was observed between drinking water inorganic arsenic and periconceptional folic acid use. As drinking water inorganic arsenic concentrations increased from 1 to 25 μg/L, the estimated protective effect of folic acid use declined (OR 0.22 to 1.03), and was not protective at higher concentrations of arsenic. No main effect of arsenic exposure on myelomeningocele risk was identified. CONCLUSIONS Our study found a significant interaction between drinking water inorganic arsenic concentration from wells used during the first trimester of pregnancy and reported intake of periconceptional folic acid supplements. Results suggest that environmental arsenic exposure reduces the effectiveness of folic acid supplementation in preventing myelomeningocele.
Collapse
Affiliation(s)
- Maitreyi Mazumdar
- Department of Neurology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, USA.
- Department of Environmental Health, Harvard School of Public Health, 665 Huntington Avenue, Boston, MA, USA.
| | | | - Rezina Hamid
- Bangladesh Medical College, 14/A Dhanmondi, Dhaka, 1209, Bangladesh.
| | - Linda Valeri
- Department of Biostatistics, Harvard School of Public Health, 655 Huntington Avenue, Boston, MA, USA.
| | - Ligi Paul
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, 711 Washington Street, Boston, MA, USA.
| | - Jacob Selhub
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, 711 Washington Street, Boston, MA, USA.
| | - Ema G Rodrigues
- Department of Neurology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, USA.
- Department of Environmental Health, Harvard School of Public Health, 665 Huntington Avenue, Boston, MA, USA.
| | - Fareesa Silva
- Department of Neurology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, USA.
| | - Selim Mia
- Dhaka Community Hospital, 190/1 Baro Moghbazar, Wireless Railgate, Dhaka, 1217, Bangladesh.
| | - Md Golam Mostofa
- Dhaka Community Hospital, 190/1 Baro Moghbazar, Wireless Railgate, Dhaka, 1217, Bangladesh.
| | - Quazi Quamruzzaman
- Dhaka Community Hospital, 190/1 Baro Moghbazar, Wireless Railgate, Dhaka, 1217, Bangladesh.
| | - Mahmuder Rahman
- Dhaka Community Hospital, 190/1 Baro Moghbazar, Wireless Railgate, Dhaka, 1217, Bangladesh.
| | - David C Christiani
- Department of Environmental Health, Harvard School of Public Health, 665 Huntington Avenue, Boston, MA, USA.
| |
Collapse
|
24
|
Wu J, Zhang C, Pei L, Chen G, Zheng X. Association between risk of birth defects occurring level and arsenic concentrations in soils of Lvliang, Shanxi province of China. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2014; 191:1-7. [PMID: 24769413 DOI: 10.1016/j.envpol.2014.04.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 03/30/2014] [Accepted: 04/03/2014] [Indexed: 06/03/2023]
Abstract
The risk of birth defects is generally accredited with genetic factors, environmental causes, but the contribution of environmental factors to birth defects is still inconclusive. With the hypothesis of associations of geochemical features distribution and birth defects risk, we collected birth records and measured the chemical components in soil samples from a high prevalence area of birth defects in Shanxi province, China. The relative risk levels among villages were estimated with conditional spatial autoregressive model and the relationships between the risk levels of the villages and the 15 types of chemical elements concentration in the cropland and woodland soils were explored. The results revealed that the arsenic levels in cropland soil showed a significant association with birth defects occurring risk in this area, which is consistent with existing evidences of arsenic as a teratogen and warrants further investigation on arsenic exposure routine to birth defect occurring risk.
Collapse
Affiliation(s)
- Jilei Wu
- Institute of Population Research, Peking University, Beijing, China.
| | - Chaosheng Zhang
- GIS Centre, Ryan Institute and School of Geography and Archaeology, National University of Ireland, Galway, Ireland
| | - Lijun Pei
- Institute of Population Research, Peking University, Beijing, China
| | - Gong Chen
- Institute of Population Research, Peking University, Beijing, China
| | - Xiaoying Zheng
- Institute of Population Research, Peking University, Beijing, China.
| |
Collapse
|
25
|
Anwar-Mohamed A, Elshenawy OH, El-Sherbeni AA, Abdelrady M, El-Kadi AO. Acute arsenic treatment alters arachidonic acid and its associated metabolite levels in the brain of C57Bl/6 mice. Can J Physiol Pharmacol 2014; 92:693-702. [DOI: 10.1139/cjpp-2014-0136] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The toxic effects of arsenic on the whole brain, as well as the discrete regions, has been previously reported for mice. We investigated the effects of acute arsenite (As(III)) on brain levels of arachidonic acid (AA) and its associated metabolites generated through cytochrome P450 (CYP), cyclooxygenase (COX), and lipoxygenase (LOX) pathways. Our results demonstrated that acute As(III) treatment (12.5 mg·(kg body mass)−1) decreases cytosolic phospholipase A2 (cPLA2) with a subsequent decrease in its catalytic activity and brain AA levels. In addition, As(III) differentially altered CYP epoxygenases and CYP ω-hydroxylases, but it did not affect brain Ephx2 mRNA or sEH catalytic activity levels. As(III)-mediated effects on Cyps caused an increase in brain 5,6-epoxyeicosatrienoic acid (5,6-EET) and 16/17-hydroxyeicosatetreinoic acid (16/17-HETE) levels, and a decrease in 18- and 20-HETE levels. Furthermore, As(III) increased cyclooxygenase-2 (COX-2) mRNA while decreasing prostaglandins F2α (PGF2α) and PGJ2. As(III) also increased brain 5-lipoxygenase (5-LOX) and 15-LOX mRNA, but decreased 12-LOX mRNA. These changes in LOX mRNA were associated with a decrease in 8/12-HETE levels only. In conclusion, this is the first demonstration that As(III) decreases AA levels coinciding with alterations to EET, HETE, and PG levels, which affects brain development and neurochemistry.
Collapse
Affiliation(s)
- Anwar Anwar-Mohamed
- Faculty of Pharmacy and Pharmaceutical Sciences, 2142J Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Osama H. Elshenawy
- Faculty of Pharmacy and Pharmaceutical Sciences, 2142J Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Ahmed A. El-Sherbeni
- Faculty of Pharmacy and Pharmaceutical Sciences, 2142J Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Mohamed Abdelrady
- Faculty of Pharmacy and Pharmaceutical Sciences, 2142J Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Ayman O.S. El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, 2142J Katz Group-Rexall Centre for Pharmacy and Health Research, University of Alberta, Edmonton, AB T6G 2E1, Canada
| |
Collapse
|
26
|
Abstract
Rates of metabolic diseases have increased at an astounding rate in recent decades. Even though poor diet and physical inactivity are central drivers, these lifestyle changes alone fail to fully account for the magnitude and rapidity of the epidemic. Thus, attention has turned to identifying novel risk factors, including the contribution of environmental endocrine disrupting chemicals. Epidemiologic and preclinical data support a role for various contaminants in the pathogenesis of diabetes. In addition to the vascular risk associated with dysglycemia, emerging evidence implicates multiple pollutants in the pathogenesis of atherosclerosis and cardiovascular disease. Reviewed herein are studies linking endocrine disruptors to these key diseases that drive significant individual and societal morbidity and mortality. Identifying chemicals associated with metabolic and cardiovascular disease as well as their mechanisms of action is critical for developing novel treatment strategies and public policy to mitigate the impact of these diseases on human health.
Collapse
Affiliation(s)
- Andrew G. Kirkley
- Committee on Molecular Pathogenesis and Molecular Medicine
- University of Chicago, Chicago, IL
| | - Robert M. Sargis
- Committee on Molecular Metabolism and Nutrition
- Kovler Diabetes Center
- Section of Endocrinology, Diabetes and Metabolism
- University of Chicago, Chicago, IL
| |
Collapse
|
27
|
Sargis RM. The hijacking of cellular signaling and the diabetes epidemic: mechanisms of environmental disruption of insulin action and glucose homeostasis. Diabetes Metab J 2014; 38:13-24. [PMID: 24627823 PMCID: PMC3950190 DOI: 10.4093/dmj.2014.38.1.13] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The burgeoning epidemic of metabolic disease causes significant societal and individual morbidity and threatens the stability of health care systems around the globe. Efforts to understand the factors that contribute to metabolic derangements are critical for reversing these troubling trends. While excess caloric consumption and physical inactivity superimposed on a susceptible genetic background are central drivers of this crisis, these factors alone fail to fully account for the magnitude and rapidity with which metabolic diseases have increased in prevalence worldwide. Recent epidemiological evidence implicates endocrine disrupting chemicals in the pathogenesis of metabolic diseases. These compounds represent a diverse array of chemicals to which humans are exposed via multiple routes in adulthood and during development. Furthermore, a growing ensemble of animal- and cell-based studies provides preclinical evidence supporting the hypothesis that environmental contaminants contribute to the development of metabolic diseases, including diabetes. Herein are reviewed studies linking specific endocrine disruptors to impairments in glucose homeostasis as well as tying these compounds to disturbances in insulin secretion and impairments in insulin signal transduction. While the data remains somewhat incomplete, the current body of evidence supports the hypothesis that our chemically polluted environment may play a contributing role in the current metabolic crisis.
Collapse
Affiliation(s)
- Robert M. Sargis
- Committee on Molecular Metabolism and Nutrition, Kovler Diabetes Center, Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
28
|
n5-STZ Diabetic Model Develops Alterations in Sciatic Nerve and Dorsal Root Ganglia Neurons of Wistar Rats. ISRN ENDOCRINOLOGY 2013; 2013:638028. [PMID: 23476801 PMCID: PMC3588209 DOI: 10.1155/2013/638028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 12/18/2012] [Indexed: 12/20/2022]
Abstract
One experimental model of diabetes mellitus (DM) similar to type
II DM, called n5-STZ, is obtained by a single injection (via i.p.)
of streptozotocin (STZ) in the 5th day of life of newborn rats.
The present investigation aimed to characterize alterations in
excitability of rat peripheral neurons in n5-STZ model. n5-STZ DM
was induced, and electrophysiological evaluation was done at 12th
week of rat life. Rats developed glucose intolerance, sensory
alteration, and hyperglycemia or near-normoglycemia (21.2 ± 1.6 and 7.4 ± 0.4 mmol/L). In near-normoglycemia group the significant
electrophysiological alteration observed was decreased in
amplitude of 2nd wave (2nd component, conduction velocity:
48.8 m/s) of compound action potential (CAP) of sciatic nerve. For
hyperglycemic rats, decreased excitability, amplitude, and
conduction velocity of 2nd CAP component of sciatic nerve were
found; a depolarization of resting potential (4-5 mV) and reduction
in maximum ascendant and descendant inclinations of action
potential were found in DRG neurons but no alteration on
Na+ current (INa+).
Thus, n5-STZ rats develop alterations in
excitability which were related to glycemic levels but were not
likely attributable to changes on INa+. Our data confirm that
n5-STZ model is a useful model to study type II DM.
Collapse
|
29
|
Hou Y, Xue P, Woods CG, Wang X, Fu J, Yarborough K, Qu W, Zhang Q, Andersen ME, Pi J. Association between arsenic suppression of adipogenesis and induction of CHOP10 via the endoplasmic reticulum stress response. ENVIRONMENTAL HEALTH PERSPECTIVES 2013; 121:237-43. [PMID: 23221991 PMCID: PMC3569692 DOI: 10.1289/ehp.1205731] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2012] [Accepted: 12/04/2012] [Indexed: 05/02/2023]
Abstract
BACKGROUND There is growing evidence that chronic exposure to inorganic arsenic (iAs) is associated with an increased prevalence of type 2 diabetes (T2D). However, the mechanisms for the diabetogenic effect of iAs are still largely unknown. White adipose tissue (WAT) actively stores and releases energy and maintains lipid and glucose homeostasis. OBJECTIVE We sought to determine the mechanisms of arsenic suppression of adipogenesis. METHODS The effects and associated mechanisms of iAs and its major metabolites on adipogenesis were determined in 3T3-L1 preadipocytes, mouse adipose-derived stromal-vascular fraction cells (ADSVFCs), and human adipose tissue-derived stem cells (ADSCs). RESULTS Exposure of 3T3-L1 preadipocytes to noncytotoxic levels of arsenic, including inorganic arsenite (iAs3+, ≤ 5 μM), inorganic arsenate (≤ 20 μM), trivalent monomethylated arsenic (MMA3+, ≤ 1 μM), and trivalent dimethylated arsenic (DMA3+, ≤ 2 μM) decreased adipogenic hormone-induced adipogenesis in a concentration-dependent manner. In addition, iAs3+, MMA3+, and DMA3+ exhibited a strong inhibitory effect on adipogenesis in primary cultured mouse ADSVFCs and human ADSCs. Time-course studies in 3T3-L1 cells revealed that inhibition of adipogenesis by arsenic occurred in the early stage of terminal adipogenic differentiation and was highly correlated with the induction of C/EBP homologous protein (CHOP10), an endoplasmic reticulum (ER) stress response protein. Induction of CHOP10 by arsenic is associated with reduced DNA-binding activity of CCAAT/enhancer-binding protein β (C/EBPβ), which regulates the transcription of peroxisome proliferator-activated receptor γ and C/EBPα. CONCLUSIONS Low-level iAs and MMA3+ trigger the ER stress response and up-regulate CHOP10, which inhibits C/EBPβ transcriptional activity, thus suppressing adipogenesis. Arsenic-induced dysfunctional adipogenesis may be associated with a reduced capacity of WAT to store lipids and with insulin resistance.
Collapse
Affiliation(s)
- Yongyong Hou
- Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina 27709, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Effects of choline on sodium arsenite-induced neural tube defects in chick embryos. Food Chem Toxicol 2012; 50:4364-74. [PMID: 22963837 DOI: 10.1016/j.fct.2012.08.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 05/16/2011] [Accepted: 06/01/2011] [Indexed: 11/20/2022]
|
31
|
Maull EA, Ahsan H, Edwards J, Longnecker MP, Navas-Acien A, Pi J, Silbergeld EK, Styblo M, Tseng CH, Thayer KA, Loomis D. Evaluation of the association between arsenic and diabetes: a National Toxicology Program workshop review. ENVIRONMENTAL HEALTH PERSPECTIVES 2012; 120:1658-70. [PMID: 22889723 PMCID: PMC3548281 DOI: 10.1289/ehp.1104579] [Citation(s) in RCA: 261] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 08/10/2012] [Indexed: 05/17/2023]
Abstract
BACKGROUND Diabetes affects an estimated 346 million persons globally, and total deaths from diabetes are projected to increase > 50% in the next decade. Understanding the role of environmental chemicals in the development or progression of diabetes is an emerging issue in environmental health. In 2011, the National Toxicology Program (NTP) organized a workshop to assess the literature for evidence of associations between certain chemicals, including inorganic arsenic, and diabetes and/or obesity to help develop a focused research agenda. This review is derived from discussions at that workshop. OBJECTIVES Our objectives were to assess the consistency, strength/weaknesses, and biological plausibility of findings in the scientific literature regarding arsenic and diabetes and to identify data gaps and areas for future evaluation or research. The extent of the existing literature was insufficient to consider obesity as an outcome. DATA SOURCES, EXTRACTION, AND SYNTHESIS Studies related to arsenic and diabetes or obesity were identified through PubMed and supplemented with relevant studies identified by reviewing the reference lists in the primary literature or review articles. CONCLUSIONS Existing human data provide limited to sufficient support for an association between arsenic and diabetes in populations with relatively high exposure levels (≥ 150 µg arsenic/L in drinking water). The evidence is insufficient to conclude that arsenic is associated with diabetes in lower exposure (< 150 µg arsenic/L drinking water), although recent studies with better measures of outcome and exposure support an association. The animal literature as a whole was inconclusive; however, studies using better measures of diabetes-relevant end points support a link between arsenic and diabetes.
Collapse
Affiliation(s)
- Elizabeth A Maull
- Biomolecular Screening Branch, Division of the National Toxicology Program, National Institute of Environmental Sciences (NIEHS), National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Parajuli RP, Fujiwara T, Umezaki M, Furusawa H, Ser PH, Watanabe C. Cord blood levels of toxic and essential trace elements and their determinants in the Terai region of Nepal: a birth cohort study. Biol Trace Elem Res 2012; 147:75-83. [PMID: 22234823 DOI: 10.1007/s12011-011-9309-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2011] [Accepted: 12/15/2011] [Indexed: 10/14/2022]
Abstract
The purpose of this study is to evaluate the cord blood level of toxic and trace elements and to identify their determinants in Terai, Nepal. One hundred pregnant women were recruited from one hospital in Chitwan, Nepal in 2008. The cord blood levels of toxic [lead (Pb), arsenic (As), and cadmium (Cd)], essential trace elements [zinc (Zn), selenium (Se), and copper (Cu)], demographic, socioeconomic, and behavioral variables were measured. The mean values of Pb, As, Cd, Zn, Se, and Cu in cord blood level were found as 31.7, 1.46, 0.39, 2,286, 175, and 667 μg/L, respectively. In the multivariate regression model, cord blood As levels from less educated mothers were higher than those from educated mothers (coefficient = -0.01, 95% confidence interval [CI] = -0.02-0.00). The maternal age was positively associated with the cord blood Cd level (coefficient = 0.02, 95% CI = 0.01-0.03), while it was negatively associated with the cord blood As level (coefficient = -0.01, 95% CI = -0.03--0.01). Cord blood levels of Pb, Zn, Se, and Cu were not associated with maternal age, socioeconomic status, living environment, and smoking status. As and Cd levels were relatively lower than those reported in previous studies in Asia, while the levels of Pb and the trace elements were similar. Less educated mothers are more likely to become a higher in utero As source to their fetus, and fetuses of older mothers were more likely to have higher in utero Cd exposure in Terai, Nepal.
Collapse
Affiliation(s)
- Rajendra Prasad Parajuli
- Department of Social Medicine, National Research Institute for Child Health and Development, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
33
|
Thayer KA, Heindel JJ, Bucher JR, Gallo MA. Role of environmental chemicals in diabetes and obesity: a National Toxicology Program workshop review. ENVIRONMENTAL HEALTH PERSPECTIVES 2012; 120:779-89. [PMID: 22296744 PMCID: PMC3385443 DOI: 10.1289/ehp.1104597] [Citation(s) in RCA: 458] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Accepted: 02/01/2012] [Indexed: 05/17/2023]
Abstract
BACKGROUND There has been increasing interest in the concept that exposures to environmental chemicals may be contributing factors to the epidemics of diabetes and obesity. On 11-13 January 2011, the National Institute of Environmental Health Sciences (NIEHS) Division of the National Toxicology Program (NTP) organized a workshop to evaluate the current state of the science on these topics of increasing public health concern. OBJECTIVE The main objective of the workshop was to develop recommendations for a research agenda after completing a critical analysis of the literature for humans and experimental animals exposed to certain environmental chemicals. The environmental exposures considered at the workshop were arsenic, persistent organic pollutants, maternal smoking/nicotine, organotins, phthalates, bisphenol A, and pesticides. High-throughput screening data from Toxicology in the 21st Century (Tox21) were also considered as a way to evaluate potential cellular pathways and generate -hypotheses for testing which and how certain chemicals might perturb biological processes related to diabetes and obesity. CONCLUSIONS Overall, the review of the existing literature identified linkages between several of the environmental exposures and type 2 diabetes. There was also support for the "developmental obesogen" hypothesis, which suggests that chemical exposures may increase the risk of obesity by altering the differentiation of adipocytes or the development of neural circuits that regulate feeding behavior. The effects may be most apparent when the developmental exposure is combined with consumption of a high-calorie, high-carbohydrate, or high-fat diet later in life. Research on environmental chemical exposures and type 1 diabetes was very limited. This lack of research was considered a critical data gap. In this workshop review, we outline the major themes that emerged from the workshop and discuss activities that NIEHS/NTP is undertaking to address research recommendations. This review also serves as an introduction to an upcoming series of articles that review the literature regarding specific exposures and outcomes in more detail.
Collapse
Affiliation(s)
- Kristina A Thayer
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, USA.
| | | | | | | |
Collapse
|
34
|
Huang CF, Chen YW, Yang CY, Tsai KS, Yang RS, Liu SH. Arsenic and diabetes: current perspectives. Kaohsiung J Med Sci 2011; 27:402-10. [PMID: 21914528 DOI: 10.1016/j.kjms.2011.05.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Accepted: 03/24/2011] [Indexed: 01/19/2023] Open
Abstract
Arsenic is a naturally occurring toxic metalloid of global concern. Many studies have indicated a dose-response relationship between accumulative arsenic exposure and the prevalence of diabetes mellitus (DM) in arseniasis-endemic areas in Taiwan and Bangladesh, where arsenic exposure occurs through drinking water. Epidemiological researches have suggested that the characteristics of arsenic-induced DM observed in arseniasis-endemic areas in Taiwan and Mexico are similar to those of non-insulin-dependent DM (Type 2 DM). These studies analyzed the association between high and chronic exposure to inorganic arsenic in drinking water and the development of DM, but the effect of exposure to low to moderate levels of inorganic arsenic on the risk of DM is unclear. Navas-Acien et al. recently proposed that a positive association existed between total urine arsenic and the prevalence of Type 2 DM in people exposed to low to moderate levels of arsenic. However, the diabetogenic role played by arsenic is still debated upon. An increase in the prevalence of DM has been observed among residents of highly arsenic-contaminated areas, whereas the findings from community-based and occupational studies in low-arsenic-exposure areas have been inconsistent. Recently, a population-based cross-sectional study showed that the current findings did not support an association between arsenic exposure from drinking water at levels less than 300 μg/L and a significantly increased risk of DM. Moreover, although the precise mechanisms for the arsenic-induced diabetogenic effect are still largely undefined, recent in vitro experimental studies indicated that inorganic arsenic or its metabolites impair insulin-dependent glucose uptake or glucose-stimulated insulin secretion. Nevertheless, the dose, the form of arsenic used, and the experimental duration in the in vivo studies varied greatly, leading to conflicting results and ambiguous interpretation of these data with respect to human exposure to arsenic in the environment. Moreover, the experimental studies were limited to the use of arsenic concentrations much higher than those relevant to human exposure. Further prospective epidemiological studies might help to clarify this controversy. The issues about environmental exposure assessment and appropriate biomarkers should also be considered. Here, we focus on the review of mechanism studies and discuss the currently available evidence and conditions for the association between environmental arsenic exposure and the development of DM.
Collapse
Affiliation(s)
- Chun Fa Huang
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | | | | | | | | | | |
Collapse
|
35
|
Song G, Cui Y, Han ZJ, Xia HF, Ma X. WITHDRAWN: Effects of choline on sodium arsenite-induced neural tube defects in chick embryos. Food Chem Toxicol 2011:S0278-6915(11)00253-5. [PMID: 21708213 DOI: 10.1016/j.fct.2011.06.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 05/16/2011] [Accepted: 06/01/2011] [Indexed: 11/19/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Ge Song
- Graduate School of Peking Union Medical College, Beijing, China; Department of Genetics, National Research Institute for Family Planning, Beijing, China
| | | | | | | | | |
Collapse
|
36
|
Oxidative stress is implicated in arsenic‐induced neural tube defects in chick embryos. Int J Dev Neurosci 2011; 29:673-80. [DOI: 10.1016/j.ijdevneu.2011.06.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 06/01/2011] [Accepted: 06/16/2011] [Indexed: 11/19/2022] Open
|
37
|
Kempson IM, Lombi E. Hair analysis as a biomonitor for toxicology, disease and health status. Chem Soc Rev 2011; 40:3915-40. [PMID: 21468435 DOI: 10.1039/c1cs15021a] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hair analysis receives a large amount of academic and commercial interest for wide-ranging applications. However, in many instances, especially for elemental or 'mineral' analysis, the degree of success of analytical interpretation has been quite minimal with respect to the extent of such endeavors. In this critical review we address the questions surrounding hair analysis with specific intent of discovering what hair concentrations can actually relate to in a biogenic sense. This is done from a chemistry perspective to explain why and how elements are incorporated into hair and their meaning. This includes an overview of variables attributed to altering hair concentrations, such as age, gender, melanin content, and other less reported factors. Hair elemental concentrations are reviewed with regard to morbidity, with specific examples of disease related effects summarized. The application of hair analysis for epidemiology and etiology studies is enforced. A section is dedicated specifically to the area of population studies with regards to mercury, which highlights how endogenous and exogenous incorporation relies on species dependant metabolism and metabolic products. Many of the considerations are relevant to other areas of interest in hair analysis, such as for drug and isotopic analysis. Inclusion of a table of elemental concentrations in hair should act as a valuable reference (298 references).
Collapse
Affiliation(s)
- Ivan M Kempson
- Institute of Physics, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 115, Taiwan.
| | | |
Collapse
|
38
|
Robinson JF, Yu X, Moreira EG, Hong S, Faustman EM. Arsenic- and cadmium-induced toxicogenomic response in mouse embryos undergoing neurulation. Toxicol Appl Pharmacol 2010; 250:117-29. [PMID: 20883709 DOI: 10.1016/j.taap.2010.09.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 09/20/2010] [Accepted: 09/22/2010] [Indexed: 01/19/2023]
Abstract
Arsenic (As) and cadmium (Cd) are well-characterized teratogens in animal models inducing embryotoxicity and neural tube defects (NTDs) when exposed during neurulation. Toxicological research is needed to resolve the specific biological processes and associated molecular pathways underlying metal-induced toxicity during this timeframe in gestational development. In this study, we investigated the dose-dependent effects of As and Cd on gene expression in C57BL/6J mouse embryos exposed in utero during neurulation (GD8) to identify significantly altered genes and corresponding biological processes associated with embryotoxicity. We quantitatively examined the toxicogenomic dose-response relationship at the gene level. Our results suggest that As and Cd induce dose-dependent gene expression alterations representing shared (cell cycle, response to UV, glutathione metabolism, RNA processing) and unique (alcohol/sugar metabolism) biological processes, which serve as robust indicators of metal-induced developmental toxicity and indicate underlying embryotoxic effects. Our observations also correlate well with previously identified impacts of As and Cd on specific genes associated with metal-induced toxicity (Cdkn1a, Mt1). In summary, we have identified in a quantitative manner As and Cd induced dose-dependent effects on gene expression in mouse embryos during a peak window of sensitivity to embryotoxicity and NTDs in the sensitive C57BL/6J strain.
Collapse
Affiliation(s)
- Joshua F Robinson
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | |
Collapse
|
39
|
Yadav S, Shi Y, Wang F, Wang H. Arsenite induces apoptosis in human mesenchymal stem cells by altering Bcl-2 family proteins and by activating intrinsic pathway. Toxicol Appl Pharmacol 2010; 244:263-72. [PMID: 20083129 DOI: 10.1016/j.taap.2010.01.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Revised: 01/04/2010] [Accepted: 01/05/2010] [Indexed: 01/23/2023]
Abstract
PURPOSE Environmental exposure to arsenic is an important public health issue. The effects of arsenic on different tissues and organs have been intensively studied. However, the effects of arsenic on bone marrow mesenchymal stem cells (MSCs) have not been reported. This study is designed to investigate the cell death process caused by arsenite and its related underlying mechanisms on MSCs. The rationale is that absorbed arsenic in the blood circulation can reach to the bone marrow and may affect the cell survival of MSCs. METHODS MSCs of passage 1 were purchased from Tulane University, grown till 70% confluency level and plated according to the experimental requirements followed by treatment with arsenite at various concentrations and time points. Arsenite (iAs(III)) induced cytotoxic effects were confirmed by cell viability and cell cycle analysis. For the presence of canonic apoptosis markers; DNA damage, exposure of intramembrane phosphotidylserine, protein and m-RNA expression levels were analyzed. RESULTS iAs(III) induced growth inhibition, G2-M arrest and apoptotic cell death in MSCs, the apoptosis induced by iAs(III) in the cultured MSCs was, via altering Bcl-2 family proteins and by involving intrinsic pathway. CONCLUSION iAs(III) can induce apoptosis in bone marrow-derived MSCs via Bcl-2 family proteins, regulating intrinsic apoptotic pathway. Due to the multipotency of MSC, acting as progenitor cells for a variety of connective tissues including bone, adipose, cartilage and muscle, these effects of arsenic may be important in assessing the health risk of the arsenic compounds and understanding the mechanisms of arsenic-induced harmful effects.
Collapse
Affiliation(s)
- Santosh Yadav
- Department of Environmental Health Sciences, School of Public Health and Tropical Medicine, Tulane University New Orleans, LA 70112, USA
| | | | | | | |
Collapse
|