1
|
Lu L, Shen L, Cui S, Huang Y, Gao Y, Zhu X, Lu S, Zhang C, Zhuang S. Angiogenic Activity and Mechanism for Bisphenols on Endothelial Cell and Mouse: Evidence of a Structural-Selective Effect. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:11803-11813. [PMID: 37505069 DOI: 10.1021/acs.est.3c03883] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Increased epidemiological evidence indicates the association of bisphenol exposure with human vascular disorders, while the underlying mechanism has not been clarified. Here, we sought to unveil the potential angiogenic effect and the underlying mechanism of bisphenols with different structural features using endothelial cells treated with an environmentally relevant concentration of bisphenols (range: 1 nM to 10 μM) and a C57BL/6 mouse model fed with doses of 0.002, 0.02, 2, and 20 mg/kg BW/day for 5 weeks. Bisphenol A (BPA) and bisphenol S (BPS) at a 1 nM level significantly increased tube formation by 45.1 and 30.2% and induced the microvessel sprouting, while tube length and microvessel sprouting were significantly inhibited by 37.2 and 55.7% after exposure to tetrabromobisphenol S (TBBPS) at 1 μM, respectively. Mechanistically, TBBPA and TBBPS significantly inhibited the interaction between phosphatidylinositol 3-kinase (PI3K) and thyroid receptor (TR), while BPA and BPS favored the interaction between PI3K and estrogen receptor (ER), resulting in abnormal PI3K signaling with consequent distinct angiogenic activity. BPA- and BPS-induced pro-angiogenic effects and TBBPS showed anti-angiogenic effects due to their distinct disruption on the TR/ER-PI3K pathway. Our work provided new evidence and mechanistic insight on the angiogenic activity of bisphenols and expanded the scope of endocrine disruptors with interference in vascular homeostasis.
Collapse
Affiliation(s)
- Liping Lu
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Lilai Shen
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Shixuan Cui
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yizhou Huang
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang, China
| | - Yuchen Gao
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiaoming Zhu
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang, China
| | - Shaoyong Lu
- Department of Pathophysiology, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Chunlong Zhang
- Department of Environmental Sciences, University of Houston-Clear Lake, 2700 Bay Area Blvd., Houston, Texas 77058, United States
| | - Shulin Zhuang
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang, China
| |
Collapse
|
2
|
Plante I, Winn LM, Vaillancourt C, Grigorova P, Parent L. Killing two birds with one stone: Pregnancy is a sensitive window for endocrine effects on both the mother and the fetus. ENVIRONMENTAL RESEARCH 2022; 205:112435. [PMID: 34843719 DOI: 10.1016/j.envres.2021.112435] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 06/13/2023]
Abstract
Pregnancy is a complex process requiring tremendous physiological changes in the mother in order to fulfill the needs of the growing fetus, and to give birth, expel the placenta and nurse the newborn. These physiological modifications are accompanied with psychological changes, as well as with variations in habits and behaviors. As a result, this period of life is considered as a sensitive window as impaired functional and physiological changes in the mother can have short- and long-term impacts on her health. In addition, dysregulation of the placenta and of mechanisms governing placentation have been linked to chronic diseases later-on in life for the fetus, in a concept known as the Developmental Origin of Health and Diseases (DOHaD). This concept stipulates that any change in the environment during the pre-conception and perinatal (in utero life and neonatal) period to puberty, can be "imprinted" in the organism, thereby impacting the health and risk of chronic diseases later in life. Pregnancy is a succession of events that is regulated, in large part, by hormones and growth factors. Therefore, small changes in hormonal balance can have important effects on both the mother and the developing fetus. An increasing number of studies demonstrate that exposure to endocrine disrupting compounds (EDCs) affect both the mother and the fetus giving rise to growing concerns surrounding these exposures. This review will give an overview of changes that happen during pregnancy with respect to the mother, the placenta, and the fetus, and of the current literature regarding the effects of EDCs during this specific sensitive window of exposure.
Collapse
Affiliation(s)
- Isabelle Plante
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada.
| | - Louise M Winn
- Queen's University, School of Environmental Studies, Department of Biomedical and Molecular Sciences, Kingston, ON, Canada
| | | | - Petya Grigorova
- Département Science et Technologie, Université TELUQ, Montreal, QC, Canada
| | - Lise Parent
- Département Science et Technologie, Université TELUQ, Montreal, QC, Canada
| |
Collapse
|
3
|
Ingaramo PI, Alarcón R, Caglieris ML, Varayoud J, Muñoz-de-Toro M, Luque EH. Altered uterine angiogenesis in rats treated with a glyphosate-based herbicide. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 296:118729. [PMID: 34953950 DOI: 10.1016/j.envpol.2021.118729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 12/02/2021] [Accepted: 12/20/2021] [Indexed: 06/14/2023]
Abstract
Glyphosate-based herbicides (GBHs) are the agrochemicals most used around the globe. However, they might have adverse effects on human and animal health. Previously, we showed that female rats neonatally exposed to GBHs exhibit altered expression of morphogenetic molecules and biomarkers of uterine development. We also observed a reduction in the size of implantation sites, altered expression of decidualization-related molecules, and increased post-implantation losses. Since decidualization comprises morphogenetic, biochemical and vascular changes, here we investigated the effects of neonatal GBH exposure on uterine angiogenesis in neonatal and pregnant rats. To achieve this, Wistar female rats were exposed to saline solution or GBH (2 mg glyphosate/kg-bw/day) on post-natal days (PND) 1, 3, 5 and 7. On PND8, uterine samples were collected for developmental studies. On PND90, the remaining females were mated and in the morning of gestational day (GD) 9, the implantation sites were collected. Angiogenesis-related molecules and cells involved in this process were identified and/or measured by immunohistochemistry or RT-PCR. On PND8, GBH-treated rats showed increased vascular endothelial growth factor (VEGF) expression and decreased Notch1, inducible nitric oxide synthase (iNOS) and Angiopoietin-2 (Ang2) mRNA levels. Vascular area, vessel diameter, endothelial cell proliferation, VEGF and Nestin protein expression, and VEGF, Notch1, iNOS and cyclooxygenase-2 (Cox-2) genes were downregulated in implantation sites of exposed females, while Ang2, VEGF receptor 1 and interleukin-10 (IL-10) were increased. Mast cells and macrophages were increased on PND8 and GD9 of treated rats. The increased Transforming growth factor-beta expression in the antimesometrial zone and IL-10 mRNA expression suggest that the M2 type is the predominant population of macrophages on implantation sites. In conclusion, neonatal GBH exposure alters the expression of angiogenesis-related molecules at neonatal uterine development and decidual reaction, suggesting altered vascular support. These alterations might contribute to the increased post-implantation losses observed in GBH-treated rats.
Collapse
Affiliation(s)
- Paola I Ingaramo
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral - Consejo Nacional de Investigaciones Científicas y Técnicas, Santa Fe, Argentina.
| | - Ramiro Alarcón
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral - Consejo Nacional de Investigaciones Científicas y Técnicas, Santa Fe, Argentina
| | - María L Caglieris
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral - Consejo Nacional de Investigaciones Científicas y Técnicas, Santa Fe, Argentina
| | - Jorgelina Varayoud
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral - Consejo Nacional de Investigaciones Científicas y Técnicas, Santa Fe, Argentina
| | - Mónica Muñoz-de-Toro
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral - Consejo Nacional de Investigaciones Científicas y Técnicas, Santa Fe, Argentina
| | - Enrique H Luque
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral - Consejo Nacional de Investigaciones Científicas y Técnicas, Santa Fe, Argentina
| |
Collapse
|
4
|
Altamirano GA, Gomez AL, Schierano-Marotti G, Muñoz-de-Toro M, Rodriguez HA, Kass L. Bisphenol A and benzophenone-3 exposure alters milk protein expression and its transcriptional regulation during functional differentiation of the mammary gland in vitro. ENVIRONMENTAL RESEARCH 2020; 191:110185. [PMID: 32946892 DOI: 10.1016/j.envres.2020.110185] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 08/03/2020] [Accepted: 08/27/2020] [Indexed: 06/11/2023]
Abstract
The plastic monomer and plasticizer bisphenol A (BPA), and the UV-filter benzophenone-3 (BP3) have been shown to have estrogenic activities that could alter mammary gland development. Our aim was to analyze whether BPA or BP3 direct exposure affects the functional differentiation of the mammary gland using an in vitro model. Mammary organoids were obtained and isolated from 8 week-old virgin female C57BL/6 mice and were differentiated on Matrigel with medium containing lactogenic hormones and exposed to: a) vehicle (0.01% ethanol); b) 1 × 10-9 M or 1 × 10-6 M BPA; or c) 1 × 10-12 M, 1 × 10-9 M or 1 × 10-6 M BP3 for 72 h. The mRNA and protein expression of estrogen receptor alpha (ESR1) and progesterone receptor (PR) were assessed. In addition, mRNA levels of PR-B isoform, glucocorticoid receptor (GR), prolactin receptor (PRLR) and Stat5a, and protein expression of pStat5a/b were evaluated at 72 h. The mRNA and protein expression of milk proteins and their DNA methylation status were also analyzed. Although mRNA level of PRLR and GR was similar between treatments, mRNA expression of ESR1, total PR, PR-B and Stat5a was increased in organoids exposed to 1 × 10-9 M BPA and 1 × 10-12 M BP3. Total PR expression was also increased with 1 × 10-6 M BPA. Nuclear ESR1 and PR expression was observed in all treated organoids; whereas nuclear pStat5a/b alveolar cells was observed only in organoids exposed to 1 × 10-9 M BPA and 1 × 10-12 M BP3. The beta-casein mRNA level was increased in both BPA concentrations and 1 × 10-12 M BP3, which was associated with hypomethylation of its promoter. The beta-casein protein expression was only increased with 1 × 10-9 M BPA or 1 × 10-12 M BP3. In contrast, BPA exposure decreased alpha-lactalbumin mRNA expression and increased DNA methylation level in different methylation-sensitive sites of the gene. Also, 1 × 10-9 M BPA decreased alpha-lactalbumin protein expression. Our results demonstrate that BPA or BP3 exposure alters milk protein synthesis and its transcriptional regulation during mammary gland differentiation in vitro.
Collapse
Affiliation(s)
- Gabriela A Altamirano
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Ayelen L Gomez
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Gonzalo Schierano-Marotti
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Mónica Muñoz-de-Toro
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Horacio A Rodriguez
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Fisiología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Laura Kass
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina.
| |
Collapse
|
5
|
Hazarika J, Ganguly M, Borgohain G, Baruah I, Sarma S, Bhuyan P, Mahanta R. Endocrine disruption: molecular interactions of chlorpyrifos and its degradation products with estrogen receptor. Struct Chem 2020. [DOI: 10.1007/s11224-020-01562-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
6
|
Zanardi MV, Schimpf MG, Gastiazoro MP, Milesi MM, Muñoz-de-Toro M, Varayoud J, Durando M. Glyphosate-based herbicide induces hyperplastic ducts in the mammary gland of aging Wistar rats. Mol Cell Endocrinol 2020; 501:110658. [PMID: 31756423 DOI: 10.1016/j.mce.2019.110658] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 11/12/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022]
Abstract
Glyphosate-based herbicide (GBH) exposure is known to have adverse effects on endocrine-related tissues. Here, we aimed to determine whether early postnatal exposure to a GBH induces long-term effects on the rat mammary gland. Thus, female Wistar pups were injected with saline solution (Control) or GBH (2 mg glyphosate/kg/day) on postnatal days (PND) 1, 3, 5 and 7. At 20 months of age, mammary gland samples were collected to determine histomorphological features, proliferation index and the expression of steroid hormone receptors expression, by immunohistochemistry, and serum samples were collected to assess 17β-estradiol (E2) and progesterone (P4) levels. GBH exposure induced morphological changes evidenced by a higher percentage of hyperplastic ducts and a fibroblastic-like stroma in the mammary gland. GBH-treated rats also showed a high expression of steroid hormone receptors in hyperplastic ducts. The results indicate that early postnatal exposure to GBH induces long-term alterations in the mammary gland morphology of aging female rats.
Collapse
Affiliation(s)
- María V Zanardi
- Instituto de Salud y Ambiente del Litoral (ISAL; UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas (FBCB), Universidad Nacional del Litoral (UNL), Santa Fe, Argentina; Cátedra de Fisiología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Marlise Guerrero Schimpf
- Instituto de Salud y Ambiente del Litoral (ISAL; UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas (FBCB), Universidad Nacional del Litoral (UNL), Santa Fe, Argentina; Cátedra de Fisiología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - María P Gastiazoro
- Instituto de Salud y Ambiente del Litoral (ISAL; UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas (FBCB), Universidad Nacional del Litoral (UNL), Santa Fe, Argentina
| | - María M Milesi
- Instituto de Salud y Ambiente del Litoral (ISAL; UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas (FBCB), Universidad Nacional del Litoral (UNL), Santa Fe, Argentina; Cátedra de Fisiología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Mónica Muñoz-de-Toro
- Instituto de Salud y Ambiente del Litoral (ISAL; UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas (FBCB), Universidad Nacional del Litoral (UNL), Santa Fe, Argentina; Cátedra de Patología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Jorgelina Varayoud
- Instituto de Salud y Ambiente del Litoral (ISAL; UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas (FBCB), Universidad Nacional del Litoral (UNL), Santa Fe, Argentina; Cátedra de Fisiología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Milena Durando
- Instituto de Salud y Ambiente del Litoral (ISAL; UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas (FBCB), Universidad Nacional del Litoral (UNL), Santa Fe, Argentina; Cátedra de Fisiología Humana, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina.
| |
Collapse
|
7
|
Leonel ECR, Campos SGP, Guerra LHA, Bedolo CM, Vilamaior PSL, Calmon MF, Rahal P, Amorim CA, Taboga SR. Impact of perinatal bisphenol A and 17β estradiol exposure: Comparing hormone receptor response. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 188:109918. [PMID: 31753310 DOI: 10.1016/j.ecoenv.2019.109918] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 10/30/2019] [Accepted: 11/02/2019] [Indexed: 06/10/2023]
Abstract
Hormonal regulation controls mammary gland (MG) development. Therefore some hormone-related factors can disrupt the early phases of MGs development, making the gland more susceptible to long term modifications in its response to circulating hormones. Endocrine disruptors, such as bisphenol A (BPA), are able to cause alterations in hormone receptor expression, leading to changes in the cell proliferation index, which may expose the tissue to neoplastic alterations. Thus, we evaluated the variations in hormone receptor expression in the MG of 6-month old Mongolian gerbils exposed to BPA and 17β estradiol during the perinatal period. Receptors for estrogen alpha (ERα), beta (ERβ), progesterone (PGR), prolactin (PRL-R), and co-localization of connexin 43 (Cx43) and ERα in gerbils were analyzed, and serum concentrations of estradiol and progesterone were assessed. No alterations in body, liver, and ovary-uterus complex weights were observed. However, there was an increase in epithelial ERα expression in the 17β estradiol (E2) group and in PGR in the BPA group. Although immunohistochemistry did not show alterations in ERβ expression, western blotting revealed a decrease in this protein in the BPA group. PRL-R was more present in epithelial cells in the vehicle control (VC), E2, and BPA groups in comparison to the intact control group. Cx43 was more frequent in E2 and BPA groups, suggesting a protective response from the gland against possible malignancy. Serum concentration of estradiol reduced in VC, E2, and BPA groups, confirming that alterations also impacts steroid levels. Consequently, perinatal exposure to BPA and the reference endogenous estrogen, 17β estradiol, are able to increase the tendency of endocrine disruption in MG in a long term manner, since repercussions are observed even 6 months after exposure.
Collapse
Affiliation(s)
- Ellen Cristina Rivas Leonel
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), Rua Cristóvão Colombo 2265, Jardim Nazareth, 15054-000, São José do Rio Preto, São Paulo, Brazil
| | - Silvana Gisele Pegorin Campos
- Federal University of São João del Rei (UFSJ), Campus Centro Oeste Dona Lindu, Avenida Sebastião Gonçalves Coelho, 400, Bairro Chanadour, 35501-296, Divinópolis, Minas Gerais, Brazil
| | - Luiz Henrique Alves Guerra
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), Rua Cristóvão Colombo 2265, Jardim Nazareth, 15054-000, São José do Rio Preto, São Paulo, Brazil
| | - Carolina Marques Bedolo
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), Rua Cristóvão Colombo 2265, Jardim Nazareth, 15054-000, São José do Rio Preto, São Paulo, Brazil
| | - Patrícia Simone Leite Vilamaior
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), Rua Cristóvão Colombo 2265, Jardim Nazareth, 15054-000, São José do Rio Preto, São Paulo, Brazil
| | - Marilia Freitas Calmon
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), Rua Cristóvão Colombo 2265, Jardim Nazareth, 15054-000, São José do Rio Preto, São Paulo, Brazil
| | - Paula Rahal
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), Rua Cristóvão Colombo 2265, Jardim Nazareth, 15054-000, São José do Rio Preto, São Paulo, Brazil
| | - Christiani Andrade Amorim
- Laboratory of Gynecology, Institute of Experimental and Clinique Research, Université Catholique de Louvain (UCL), Avenue Mounier 52, Bte B1.52.02, 1200, Brussels, Belgium
| | - Sebastião Roberto Taboga
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), Rua Cristóvão Colombo 2265, Jardim Nazareth, 15054-000, São José do Rio Preto, São Paulo, Brazil.
| |
Collapse
|
8
|
Renaud L, Huff M, da Silveira WA, Angert M, Haas M, Hardiman G. Genome-Wide Analysis of Low Dose Bisphenol-A (BPA) Exposure in Human Prostate Cells. Curr Genomics 2019; 20:260-274. [PMID: 32030086 PMCID: PMC6983955 DOI: 10.2174/1389202920666190603123040] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 02/07/2023] Open
Abstract
Endocrine disrupting compounds (EDCs) have the potential to cause adverse effects on wild-life and human health. Two important EDCs are the synthetic estrogen 17α-ethynylestradiol (EE2) and bisphenol-A (BPA) both of which are xenoestrogens (XEs) as they bind the estrogen receptor and dis-rupt estrogen physiology in mammals and other vertebrates. In the recent years the influence of XEs on oncogenes, specifically in relation to breast and prostate cancer has been the subject of considerable study.
Collapse
Affiliation(s)
- Ludivine Renaud
- 1Department of Medicine, Medical University of South Carolina, Charleston, SC, USA; 2MUSC Bioinformatics, Center for Genomic Medicine, Medical University of South Carolina, Charleston, SC, USA; 3MS in Biomedical Sciences Program, Medical University of South Carolina, Charleston, SC, USA; 4School of Biological Sciences and Institute for Global Food Security, Queens University Belfast, BelfastBT9 5AG, UK; 5Department of Medicine, University of California, La Jolla, CA, USA; 6Moores UCSD Cancer Center, University of California San Diego, La Jolla, California, CA, USA; 7Division of Biological Sciences, University of California San Diego, La Jolla, California, CA, USA
| | - Matthew Huff
- 1Department of Medicine, Medical University of South Carolina, Charleston, SC, USA; 2MUSC Bioinformatics, Center for Genomic Medicine, Medical University of South Carolina, Charleston, SC, USA; 3MS in Biomedical Sciences Program, Medical University of South Carolina, Charleston, SC, USA; 4School of Biological Sciences and Institute for Global Food Security, Queens University Belfast, BelfastBT9 5AG, UK; 5Department of Medicine, University of California, La Jolla, CA, USA; 6Moores UCSD Cancer Center, University of California San Diego, La Jolla, California, CA, USA; 7Division of Biological Sciences, University of California San Diego, La Jolla, California, CA, USA
| | - Willian A da Silveira
- 1Department of Medicine, Medical University of South Carolina, Charleston, SC, USA; 2MUSC Bioinformatics, Center for Genomic Medicine, Medical University of South Carolina, Charleston, SC, USA; 3MS in Biomedical Sciences Program, Medical University of South Carolina, Charleston, SC, USA; 4School of Biological Sciences and Institute for Global Food Security, Queens University Belfast, BelfastBT9 5AG, UK; 5Department of Medicine, University of California, La Jolla, CA, USA; 6Moores UCSD Cancer Center, University of California San Diego, La Jolla, California, CA, USA; 7Division of Biological Sciences, University of California San Diego, La Jolla, California, CA, USA
| | - Mila Angert
- 1Department of Medicine, Medical University of South Carolina, Charleston, SC, USA; 2MUSC Bioinformatics, Center for Genomic Medicine, Medical University of South Carolina, Charleston, SC, USA; 3MS in Biomedical Sciences Program, Medical University of South Carolina, Charleston, SC, USA; 4School of Biological Sciences and Institute for Global Food Security, Queens University Belfast, BelfastBT9 5AG, UK; 5Department of Medicine, University of California, La Jolla, CA, USA; 6Moores UCSD Cancer Center, University of California San Diego, La Jolla, California, CA, USA; 7Division of Biological Sciences, University of California San Diego, La Jolla, California, CA, USA
| | - Martin Haas
- 1Department of Medicine, Medical University of South Carolina, Charleston, SC, USA; 2MUSC Bioinformatics, Center for Genomic Medicine, Medical University of South Carolina, Charleston, SC, USA; 3MS in Biomedical Sciences Program, Medical University of South Carolina, Charleston, SC, USA; 4School of Biological Sciences and Institute for Global Food Security, Queens University Belfast, BelfastBT9 5AG, UK; 5Department of Medicine, University of California, La Jolla, CA, USA; 6Moores UCSD Cancer Center, University of California San Diego, La Jolla, California, CA, USA; 7Division of Biological Sciences, University of California San Diego, La Jolla, California, CA, USA
| | - Gary Hardiman
- 1Department of Medicine, Medical University of South Carolina, Charleston, SC, USA; 2MUSC Bioinformatics, Center for Genomic Medicine, Medical University of South Carolina, Charleston, SC, USA; 3MS in Biomedical Sciences Program, Medical University of South Carolina, Charleston, SC, USA; 4School of Biological Sciences and Institute for Global Food Security, Queens University Belfast, BelfastBT9 5AG, UK; 5Department of Medicine, University of California, La Jolla, CA, USA; 6Moores UCSD Cancer Center, University of California San Diego, La Jolla, California, CA, USA; 7Division of Biological Sciences, University of California San Diego, La Jolla, California, CA, USA
| |
Collapse
|
9
|
Grasselli F, Bussolati S, Ramoni R, Grolli S, Basini G. Simazine, a triazine herbicide, disrupts swine granulosa cell functions. Anim Reprod 2018; 15:3-11. [PMID: 33365088 PMCID: PMC7746213 DOI: 10.21451/1984-3143-2017-ar960] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The triazine herbicide simazine is a pesticide commonly detected in surface and ground waters,
although banned in most European countries since 2004. Concerns for humans and animal health
result from its potential endocrine disrupting action, that can lead to reproductive disorders.
The present in vitro study was undertaken to study simazine effects on
swine granulosa cell function, namely cell viability, proliferation, steroidogenesis
and NO production. Moreover, the ability of this substance to interfere with the angiogenetic
process, a crucial event in reproductive function, was taken into account. Our data document
that simazine treatment, at 0.1 or 10 μM concentration levels, stimulates granulosa
cell proliferation and viability and impairs steroidogenesis, increasing in particular
progesterone production. In addition, the in vitro angiogenesis bioassay
revealed a significant simazine stimulatory effect on immortalized porcine Aortic Endothelial
Cell proliferation. Collectively, these results show that simazine can display disruptive
effects on ovarian cell functional parameters, possibly resulting in reproductive dysfunction.
This hypothesis is also supported by the observed pro-angiogenetic properties of this herbicide,
as already suggested for different endocrine disruptors.
Collapse
|
10
|
Li S, Wang B, Tang Q, Liu J, Yang X. Bisphenol A triggers proliferation and migration of laryngeal squamous cell carcinoma via GPER mediated upregulation of IL-6. Cell Biochem Funct 2017; 35:209-216. [PMID: 28466560 DOI: 10.1002/cbf.3265] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 03/13/2017] [Accepted: 03/14/2017] [Indexed: 12/14/2022]
Abstract
Bisphenol A (BPA) can be accumulated into the human body via food intake and inhalation. Numerous studies indicated that BPA can trigger the tumorigenesis and progression of cancer cells. Laryngeal cancer cells can be exposed to BPA directly via food digestion, while there were very limited data concerning the effect of BPA on the development of laryngeal squamous cell carcinoma (LSCC). Our present study revealed that nanomolar BPA can trigger the proliferation of LSCC cells. Bisphenol A also increased the in vitro migration and invasion of LSCC cells and upregulated the expression of matrix metallopeptidase 2. Among various chemokines tested, the expression of IL-6 was significantly increased in LSCC cells treated with BPA for 24 hours. Neutralization antibody of IL-6 or si-IL-6 can attenuate BPA-induced proliferation and migration of LSCC cells. Targeted inhibition of G protein-coupled estrogen receptor, while not estrogen receptor (ERα), abolished BPA-induced IL-6 expression, proliferation, and migration of LSCC cells. The increased IL-6 can further activate its downstream signal molecule STAT3, which was evidenced by the results of increased phosphorylation and nuclear translocation of STAT3, while si-IL-6 and si-GPER can both reverse BPA-induced activation of STAT3. Collectively, our present study revealed that BPA can trigger the progression of LSCC via GPER-mediated upregulation of IL-6. Therefore, more attention should be paid for the BPA exposure on the development of laryngeal cancer.
Collapse
Affiliation(s)
- Shisheng Li
- Department of Otolaryngology, Head and Neck Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Bin Wang
- Department of Otolaryngology, Head and Neck Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Qinglai Tang
- Department of Otolaryngology, Head and Neck Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Jiajia Liu
- Department of Otolaryngology, Head and Neck Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Xinming Yang
- Department of Otolaryngology, Head and Neck Surgery, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| |
Collapse
|
11
|
Perinatal Exposure to Bisphenol A or Diethylstilbestrol Increases the Susceptibility to Develop Mammary Gland Lesions After Estrogen Replacement Therapy in Middle-Aged Rats. Discov Oncol 2017; 8:78-89. [DOI: 10.1007/s12672-016-0282-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 12/23/2016] [Indexed: 11/26/2022] Open
|
12
|
Kobayashi K, Matsuyama W, Arai Y, Koizumi S, Shimizu T, Tomioka R, Sasaki K. Boiogito Increases the Metabolism of Fatty Acids in Proximal Tubular Cells through Peroxisome Proliferators-Activated Receptor (PPAR) α Agonistic Activity. Biol Pharm Bull 2016; 39:143-7. [PMID: 26725438 DOI: 10.1248/bpb.b15-00678] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The promotion of fatty acid metabolism, to which peroxisome proliferators-activated receptor (PPAR) α contributes, has been suggested to participate in maintaining the function of renal proximal tubular epithelial cells (PTECs). The loading of fatty acids to PTECs could result in cell inflammation and cell death. A "Kampo" medicine, Boiogito (BO), is used to treat overweight women exhibiting chronic fatigue and edema in the lower extremities or knees. BO improves renal function by reducing the portion of fatty acids, thereby preventing damage to PTECs. In this study, BO and Astragalus Root (AsR), a constituent crude drug of BO, were administered orally to intravenously bovine serum albumin (BSA)-administered mice to evaluate the PPARα-cAMP responsive element binding protein (CREB) binding protein (CBP) complex binding activity and/or mRNA expression of PPARα, as quantified by enzyme-linked immunosorbent assay (ELISA) and/or polymerase chain reaction (PCR). Increases in PPARα-CBP complex binding activity and the expression of PPARα mRNA were observed not only in BO-administered mice but also in AsR-administered mice, accompanied by a decrease in the amount of renal fatty acid.
Collapse
Affiliation(s)
- Kyoko Kobayashi
- Department of Pharmacognosy, Tohoku Pharmaceutical University
| | | | | | | | | | | | | |
Collapse
|
13
|
Belair DG, Schwartz MP, Knudsen T, Murphy WL. Human iPSC-derived endothelial cell sprouting assay in synthetic hydrogel arrays. Acta Biomater 2016; 39:12-24. [PMID: 27181878 PMCID: PMC5228278 DOI: 10.1016/j.actbio.2016.05.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/10/2016] [Accepted: 05/11/2016] [Indexed: 01/09/2023]
Abstract
UNLABELLED Activation of vascular endothelial cells (ECs) by growth factors initiates a cascade of events during angiogenesis in vivo consisting of EC tip cell selection, sprout formation, EC stalk cell proliferation, and ultimately vascular stabilization by support cells. Although EC functional assays can recapitulate one or more aspects of angiogenesis in vitro, they are often limited by undefined substrates and lack of dependence on key angiogenic signaling axes. Here, we designed and characterized a chemically-defined model of endothelial sprouting behavior in vitro using human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs). We rapidly encapsulated iPSC-ECs at high density in poly(ethylene glycol) (PEG) hydrogel spheres using thiol-ene chemistry and subsequently encapsulated cell-dense hydrogel spheres in a cell-free hydrogel layer. The hydrogel sprouting array supported pro-angiogenic phenotype of iPSC-ECs and supported growth factor-dependent proliferation and sprouting behavior. iPSC-ECs in the sprouting model responded appropriately to several reference pharmacological angiogenesis inhibitors of vascular endothelial growth factor, NF-κB, matrix metalloproteinase-2/9, protein kinase activity, and β-tubulin, which confirms their functional role in endothelial sprouting. A blinded screen of 38 putative vascular disrupting compounds from the US Environmental Protection Agency's ToxCast library identified six compounds that inhibited iPSC-EC sprouting and five compounds that were overtly cytotoxic to iPSC-ECs at a single concentration. The chemically-defined iPSC-EC sprouting model (iSM) is thus amenable to enhanced-throughput screening of small molecular libraries for effects on angiogenic sprouting and iPSC-EC toxicity assessment. STATEMENT OF SIGNIFICANCE Angiogenesis assays that are commonly used for drug screening and toxicity assessment applications typically utilize natural substrates like Matrigel(TM) that are difficult to spatially pattern, costly, ill-defined, and may exhibit lot-to-lot variability. Herein, we describe a novel angiogenic sprouting assay using chemically-defined, bioinert poly(ethylene glycol) hydrogels functionalized with biomimetic peptides to promote cell attachment and degradation in a reproducible format that may mitigate the need for natural substrates. The quantitative assay of angiogenic sprouting here enables precise control over the initial conditions and can be formulated into arrays for screening. The sprouting assay here was dependent on key angiogenic signaling axes in a screen of angiogenesis inhibitors and a blinded screen of putative vascular disrupting compounds from the US-EPA.
Collapse
Affiliation(s)
- David G Belair
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael P Schwartz
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Thomas Knudsen
- National Center for Computational Toxicology, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC, USA
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA; Material Science Program, University of Wisconsin-Madison, Madison, WI, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
14
|
Mandrup K, Boberg J, Isling LK, Christiansen S, Hass U. Low-dose effects of bisphenol A on mammary gland development in rats. Andrology 2016; 4:673-83. [PMID: 27088260 DOI: 10.1111/andr.12193] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 02/16/2016] [Accepted: 03/01/2016] [Indexed: 12/19/2022]
Abstract
Bisphenol A (BPA) is widely used in food contact materials, toys, and other products. Several studies have indicated that effects observed at doses near human exposure levels may not be observed at higher doses. Many studies have shown effects on mammary glands at low doses of BPA, however, because of small number of animals or few doses investigated these data have not been used by EFSA as point of departure for the newly assessed tolerable daily intake (TDI). We performed a study with perinatal exposure to BPA (0, 0.025, 0.25, 5, and 50 mg/kg bw/day) in rats (n = 22 mated/group). One of the aims was to perform a study robust enough to contribute to the risk assessment of BPA and to elucidate possible biphasic dose-response relationships. We investigated mammary gland effects in the offspring at 22, 100, and 400 days of age. Male offspring showed increased mammary outgrowth on pup day (PD) 22 at 0.025 mg/kg BPA, indicating an increased mammary development at this low dose only. Increased prevalence of intraductal hyperplasia was observed in BPA females exposed to 0.25 mg/kg at PD 400, but not at PD 100, and not at higher or lower doses. The present findings support data from the published literature showing that perinatal exposure to BPA can induce increased mammary growth and proliferative lesions in rodents. Our results indicate that low-dose exposure to BPA can affect mammary gland development in male and female rats, although higher doses show a different pattern of effects. The observed intraductal hyperplasia in female rats could be associated with an increased risk for developing hyperplastic lesions, which are parallels to early signs of breast neoplasia in women. Collectively, current knowledge on effects of BPA on mammary gland at low doses indicates that highly exposed humans may not be sufficiently protected.
Collapse
Affiliation(s)
- K Mandrup
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Søborg, Denmark
| | - J Boberg
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Søborg, Denmark
| | - L K Isling
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Søborg, Denmark
| | - S Christiansen
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Søborg, Denmark
| | - U Hass
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Søborg, Denmark
| |
Collapse
|
15
|
Ventura C, Nieto MRR, Bourguignon N, Lux-Lantos V, Rodriguez H, Cao G, Randi A, Cocca C, Núñez M. Pesticide chlorpyrifos acts as an endocrine disruptor in adult rats causing changes in mammary gland and hormonal balance. J Steroid Biochem Mol Biol 2016; 156:1-9. [PMID: 26518068 DOI: 10.1016/j.jsbmb.2015.10.010] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 10/02/2015] [Accepted: 10/09/2015] [Indexed: 11/30/2022]
Abstract
Endocrine disruptors (EDs) are compounds that interfere with hormone regulation and influence mammary carcinogenesis. We have previously demonstrated that the pesticide chlorpyrifos (CPF) acts as an ED in vitro, since it induces human breast cancer cells proliferation through estrogen receptor alpha (ERα) pathway. In this work, we studied the effects of CPF at environmental doses (0.01 and 1mg/kg/day) on mammary gland, steroid hormone receptors expression and serum steroid hormone levels. It was carried out using female Sprague-Dawley 40-days-old rats exposed to the pesticide during 100 days. We observed a proliferating ductal network with a higher number of ducts and alveolar structures. We also found an increased number of benign breast diseases, such as hyperplasia and adenosis. CPF enhanced progesterone receptor (PgR) along with the proliferating cell nuclear antigen (PCNA) in epithelial ductal cells. On the other hand, the pesticide reduced the expression of co-repressors of estrogen receptor activity REA and SMRT and it decreased serum estradiol (E2), progesterone (Pg) and luteinizing hormone (LH) levels. Finally, we found a persistent decrease in LH levels among ovariectomized rats exposed to CPF. Therefore, CPF alters the endocrine balance acting as an ED in vivo. These findings warn about the harmful effects that CPF exerts on mammary gland, suggesting that this compound may act as a risk factor for breast cancer.
Collapse
Affiliation(s)
- Clara Ventura
- Laboratorio de Radioisótopos, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
| | - María Rosa Ramos Nieto
- Laboratorio de Radioisótopos, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
| | - Nadia Bourguignon
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental (IByME), CONICET, Argentina
| | - Victoria Lux-Lantos
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental (IByME), CONICET, Argentina
| | - Horacio Rodriguez
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Argentina
| | - Gabriel Cao
- Instituto de Investigaciones Cardiológicas, CONICET, Argentina
| | - Andrea Randi
- Laboratorio de Efectos Biológicos de Contaminantes Ambientales, Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Claudia Cocca
- Laboratorio de Radioisótopos, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
| | - Mariel Núñez
- Laboratorio de Radioisótopos, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina.
| |
Collapse
|
16
|
Durando M, Canesini G, Cocito LL, Galoppo GH, Zayas MA, Luque EH, Muñoz-de-Toro M. Histomorphological changes in testes of broad-snouted caimans (Caiman latirostris) associated within ovoexposure to endocrine-disrupting chemicals. ACTA ACUST UNITED AC 2015; 325:84-96. [DOI: 10.1002/jez.1999] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 11/10/2015] [Accepted: 11/16/2015] [Indexed: 12/25/2022]
Affiliation(s)
- Milena Durando
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas; Universidad Nacional del Litoral (UNL); Santa Fe Argentina
| | - Guillermina Canesini
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas; Universidad Nacional del Litoral (UNL); Santa Fe Argentina
| | - Laura L. Cocito
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas; Universidad Nacional del Litoral (UNL); Santa Fe Argentina
| | - Germán H. Galoppo
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas; Universidad Nacional del Litoral (UNL); Santa Fe Argentina
| | - Marcelo A. Zayas
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas; Universidad Nacional del Litoral (UNL); Santa Fe Argentina
| | - Enrique H. Luque
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas; Universidad Nacional del Litoral (UNL); Santa Fe Argentina
| | - Mónica Muñoz-de-Toro
- Instituto de Salud y Ambiente del Litoral (ISAL, UNL-CONICET), Facultad de Bioquímica y Ciencias Biológicas; Universidad Nacional del Litoral (UNL); Santa Fe Argentina
| |
Collapse
|
17
|
Hexachlorobenzene promotes angiogenesis in vivo, in a breast cancer model and neovasculogenesis in vitro, in the human microvascular endothelial cell line HMEC-1. Toxicol Lett 2015; 239:53-64. [DOI: 10.1016/j.toxlet.2015.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 08/31/2015] [Accepted: 09/03/2015] [Indexed: 12/13/2022]
|
18
|
Ochieng J, Nangami GN, Ogunkua O, Miousse IR, Koturbash I, Odero-Marah V, McCawley LJ, Nangia-Makker P, Ahmed N, Luqmani Y, Chen Z, Papagerakis S, Wolf GT, Dong C, Zhou BP, Brown DG, Colacci AM, Hamid RA, Mondello C, Raju J, Ryan EP, Woodrick J, Scovassi AI, Singh N, Vaccari M, Roy R, Forte S, Memeo L, Salem HK, Amedei A, Al-Temaimi R, Al-Mulla F, Bisson WH, Eltom SE. The impact of low-dose carcinogens and environmental disruptors on tissue invasion and metastasis. Carcinogenesis 2015; 36 Suppl 1:S128-59. [PMID: 26106135 DOI: 10.1093/carcin/bgv034] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The purpose of this review is to stimulate new ideas regarding low-dose environmental mixtures and carcinogens and their potential to promote invasion and metastasis. Whereas a number of chapters in this review are devoted to the role of low-dose environmental mixtures and carcinogens in the promotion of invasion and metastasis in specific tumors such as breast and prostate, the overarching theme is the role of low-dose carcinogens in the progression of cancer stem cells. It is becoming clearer that cancer stem cells in a tumor are the ones that assume invasive properties and colonize distant organs. Therefore, low-dose contaminants that trigger epithelial-mesenchymal transition, for example, in these cells are of particular interest in this review. This we hope will lead to the collaboration between scientists who have dedicated their professional life to the study of carcinogens and those whose interests are exclusively in the arena of tissue invasion and metastasis.
Collapse
Affiliation(s)
- Josiah Ochieng
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA, Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA, Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA, Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA, Department of Pathology, Wayne State University, Detroit, MI 48201, USA, Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia, Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA, Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA, Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia, Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy, Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA, Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India, Mediterranean Institute of Oncology, Viagrande 95029, Italy, Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt, Department of Experimental and
| | - Gladys N Nangami
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA, Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA, Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA, Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA, Department of Pathology, Wayne State University, Detroit, MI 48201, USA, Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia, Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA, Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA, Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia, Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy, Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA, Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India, Mediterranean Institute of Oncology, Viagrande 95029, Italy, Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt, Department of Experimental and
| | - Olugbemiga Ogunkua
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA, Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA, Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA, Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA, Department of Pathology, Wayne State University, Detroit, MI 48201, USA, Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia, Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA, Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA, Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia, Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy, Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA, Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India, Mediterranean Institute of Oncology, Viagrande 95029, Italy, Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt, Department of Experimental and
| | - Isabelle R Miousse
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Valerie Odero-Marah
- Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
| | - Lisa J McCawley
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA
| | | | - Nuzhat Ahmed
- Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia
| | - Yunus Luqmani
- Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | - Zhenbang Chen
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA, Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA, Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA, Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA, Department of Pathology, Wayne State University, Detroit, MI 48201, USA, Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia, Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA, Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA, Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia, Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy, Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA, Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India, Mediterranean Institute of Oncology, Viagrande 95029, Italy, Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt, Department of Experimental and
| | - Silvana Papagerakis
- Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA
| | - Gregory T Wolf
- Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA
| | - Chenfang Dong
- Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Binhua P Zhou
- Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA
| | - Dustin G Brown
- Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Anna Maria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Roslida A Hamid
- Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia
| | - Chiara Mondello
- Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy
| | - Jayadev Raju
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Jordan Woodrick
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - A Ivana Scovassi
- Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy
| | - Neetu Singh
- Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Rabindra Roy
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Stefano Forte
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Hosni K Salem
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze 50134, Italy and
| | - Rabeah Al-Temaimi
- Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | - Fahd Al-Mulla
- Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | - William H Bisson
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA
| | - Sakina E Eltom
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA, Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA, Department of Biology/Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA, Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA, Department of Pathology, Wayne State University, Detroit, MI 48201, USA, Department of Obstetrics and Gynecology, University of Melbourne, Melbourne, Victoria, Australia, Faculty of Pharmacy, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Otolaryngology, University of Michigan Medical College, Ann Arbor, MI 48109, USA, Department of Molecular & Cellular Biochemistry, University of Kentucky, Lexington, KY 40506, USA, Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia, Istituto di Genetica Molecolare, CNR, via Abbiategrasso 207, 27100 Pavia, Italy, Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA, Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India, Mediterranean Institute of Oncology, Viagrande 95029, Italy, Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt, Department of Experimental and
| |
Collapse
|
19
|
Narayanan KB, Ali M, Barclay BJ, Cheng QS, D'Abronzo L, Dornetshuber-Fleiss R, Ghosh PM, Gonzalez Guzman MJ, Lee TJ, Leung PS, Li L, Luanpitpong S, Ratovitski E, Rojanasakul Y, Romano MF, Romano S, Sinha RK, Yedjou C, Al-Mulla F, Al-Temaimi R, Amedei A, Brown DG, Ryan EP, Colacci A, Hamid RA, Mondello C, Raju J, Salem HK, Woodrick J, Scovassi AI, Singh N, Vaccari M, Roy R, Forte S, Memeo L, Kim SY, Bisson WH, Lowe L, Park HH. Disruptive environmental chemicals and cellular mechanisms that confer resistance to cell death. Carcinogenesis 2015; 36 Suppl 1:S89-110. [PMID: 26106145 DOI: 10.1093/carcin/bgv032] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cell death is a process of dying within biological cells that are ceasing to function. This process is essential in regulating organism development, tissue homeostasis, and to eliminate cells in the body that are irreparably damaged. In general, dysfunction in normal cellular death is tightly linked to cancer progression. Specifically, the up-regulation of pro-survival factors, including oncogenic factors and antiapoptotic signaling pathways, and the down-regulation of pro-apoptotic factors, including tumor suppressive factors, confers resistance to cell death in tumor cells, which supports the emergence of a fully immortalized cellular phenotype. This review considers the potential relevance of ubiquitous environmental chemical exposures that have been shown to disrupt key pathways and mechanisms associated with this sort of dysfunction. Specifically, bisphenol A, chlorothalonil, dibutyl phthalate, dichlorvos, lindane, linuron, methoxychlor and oxyfluorfen are discussed as prototypical chemical disruptors; as their effects relate to resistance to cell death, as constituents within environmental mixtures and as potential contributors to environmental carcinogenesis.
Collapse
Affiliation(s)
- Kannan Badri Narayanan
- Department of Chemistry and Biochemistry, Yeungnam University, Gyeongsan 712-749, South Korea, Sultan Zainal Abidin University, Malaysia, Plant Biotechnologies Inc, St. Albert AB, Canada, Computer Science Department, Southern Illinois University, Carbondale, IL 62901, USA, Department of Urology, University of California Davis, Sacramento, CA 95817, USA, Department of Pharmacology and Toxicology, University of Vienna, Austria, University of Puerto Rico, Medical Sciences Campus, School of Public Health, Nutrition Program, San Juan Puerto Rico 00936-5067, USA, Department of Anatomy, College of Medicine, Yeungnam University, Daegu, 705-717, South Korea, School of Biomedical Science, The Chinese University Of Hong Kong, Hong Kong, China, Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand, Department of Otolaryngology/Head and Neck Surgery, Head and Neck Cancer Research Division, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA, Department of Pharmaceutical Sciences, Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506, USA, Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, 80131 Naples, Italy, Department of Molecular and Experimental Medicine, MEM 180, The Scripps Research Institute, La Jolla, CA 92037, USA, Department of Biology, Jackson State University, Jackson, MS 39217, USA, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Experimental and Clinical Medicine, University of Firenze, Firenze, 50134, Italy, Department of Environmental and Radiological Health Sciences, Colorado state University/ Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna, 40126, Italy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Se
| | - Manaf Ali
- Sultan Zainal Abidin University, Malaysia
| | | | - Qiang Shawn Cheng
- Computer Science Department, Southern Illinois University, Carbondale, IL 62901, USA
| | - Leandro D'Abronzo
- Department of Urology, University of California Davis, Sacramento, CA 95817, USA
| | | | - Paramita M Ghosh
- Department of Urology, University of California Davis, Sacramento, CA 95817, USA
| | - Michael J Gonzalez Guzman
- University of Puerto Rico, Medical Sciences Campus, School of Public Health, Nutrition Program, San Juan Puerto Rico 00936-5067, USA
| | - Tae-Jin Lee
- Department of Anatomy, College of Medicine, Yeungnam University, Daegu, 705-717, South Korea
| | - Po Sing Leung
- School of Biomedical Science, The Chinese University Of Hong Kong, Hong Kong, China
| | - Lin Li
- School of Biomedical Science, The Chinese University Of Hong Kong, Hong Kong, China
| | - Suidjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Edward Ratovitski
- Department of Otolaryngology/Head and Neck Surgery, Head and Neck Cancer Research Division, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yon Rojanasakul
- Department of Pharmaceutical Sciences, Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506, USA
| | - Maria Fiammetta Romano
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, 80131 Naples, Italy
| | - Simona Romano
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, 80131 Naples, Italy
| | - Ranjeet K Sinha
- Department of Molecular and Experimental Medicine, MEM 180, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Clement Yedjou
- Department of Biology, Jackson State University, Jackson, MS 39217, USA
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | | | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, 50134, Italy
| | - Dustin G Brown
- Department of Environmental and Radiological Health Sciences, Colorado state University/ Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences, Colorado state University/ Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Annamaria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna, 40126, Italy
| | - Roslida A Hamid
- Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor 43400, Malaysia
| | - Chiara Mondello
- Institute of Molecular Genetics, National Research Council, Pavia, 27100, Italy
| | - Jayadev Raju
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario, K1A0K9, Canada
| | - Hosni K Salem
- Urology Department, Kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo, 12515, Egypt
| | - Jordan Woodrick
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, 20057, USA
| | - A Ivana Scovassi
- Institute of Molecular Genetics, National Research Council, Pavia, 27100, Italy
| | - Neetu Singh
- Advenced Molecular Science Research Centre, King George's Medical University, Lucknow, Uttar Pradesh, 226003, India
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna, 40126, Italy
| | - Rabindra Roy
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, 20057, USA
| | - Stefano Forte
- Mediterranean Institute of Oncology, Viagrande, 95029, Italy
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, Viagrande, 95029, Italy
| | - Seo Yun Kim
- Department of Internal Medicine, Korea Cancer Center Hospital, Seoul 139-706, South Korea
| | - William H Bisson
- Environmental and Molecular Toxicology, Environmental Health Science Center, Oregon State University, Corvallis, OR 97331, USA and
| | - Leroy Lowe
- Getting to Know Cancer, Truro, Nova Scotia, Canada
| | - Hyun Ho Park
- Department of Chemistry and Biochemistry, Yeungnam University, Gyeongsan 712-749, South Korea, Sultan Zainal Abidin University, Malaysia, Plant Biotechnologies Inc, St. Albert AB, Canada, Computer Science Department, Southern Illinois University, Carbondale, IL 62901, USA, Department of Urology, University of California Davis, Sacramento, CA 95817, USA, Department of Pharmacology and Toxicology, University of Vienna, Austria, University of Puerto Rico, Medical Sciences Campus, School of Public Health, Nutrition Program, San Juan Puerto Rico 00936-5067, USA, Department of Anatomy, College of Medicine, Yeungnam University, Daegu, 705-717, South Korea, School of Biomedical Science, The Chinese University Of Hong Kong, Hong Kong, China, Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand, Department of Otolaryngology/Head and Neck Surgery, Head and Neck Cancer Research Division, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA, Department of Pharmaceutical Sciences, Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506, USA, Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, 80131 Naples, Italy, Department of Molecular and Experimental Medicine, MEM 180, The Scripps Research Institute, La Jolla, CA 92037, USA, Department of Biology, Jackson State University, Jackson, MS 39217, USA, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Experimental and Clinical Medicine, University of Firenze, Firenze, 50134, Italy, Department of Environmental and Radiological Health Sciences, Colorado state University/ Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna, 40126, Italy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Se
| |
Collapse
|
20
|
Langie SAS, Koppen G, Desaulniers D, Al-Mulla F, Al-Temaimi R, Amedei A, Azqueta A, Bisson WH, Brown DG, Brunborg G, Charles AK, Chen T, Colacci A, Darroudi F, Forte S, Gonzalez L, Hamid RA, Knudsen LE, Leyns L, Lopez de Cerain Salsamendi A, Memeo L, Mondello C, Mothersill C, Olsen AK, Pavanello S, Raju J, Rojas E, Roy R, Ryan EP, Ostrosky-Wegman P, Salem HK, Scovassi AI, Singh N, Vaccari M, Van Schooten FJ, Valverde M, Woodrick J, Zhang L, van Larebeke N, Kirsch-Volders M, Collins AR. Causes of genome instability: the effect of low dose chemical exposures in modern society. Carcinogenesis 2015; 36 Suppl 1:S61-88. [PMID: 26106144 DOI: 10.1093/carcin/bgv031] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Genome instability is a prerequisite for the development of cancer. It occurs when genome maintenance systems fail to safeguard the genome's integrity, whether as a consequence of inherited defects or induced via exposure to environmental agents (chemicals, biological agents and radiation). Thus, genome instability can be defined as an enhanced tendency for the genome to acquire mutations; ranging from changes to the nucleotide sequence to chromosomal gain, rearrangements or loss. This review raises the hypothesis that in addition to known human carcinogens, exposure to low dose of other chemicals present in our modern society could contribute to carcinogenesis by indirectly affecting genome stability. The selected chemicals with their mechanisms of action proposed to indirectly contribute to genome instability are: heavy metals (DNA repair, epigenetic modification, DNA damage signaling, telomere length), acrylamide (DNA repair, chromosome segregation), bisphenol A (epigenetic modification, DNA damage signaling, mitochondrial function, chromosome segregation), benomyl (chromosome segregation), quinones (epigenetic modification) and nano-sized particles (epigenetic pathways, mitochondrial function, chromosome segregation, telomere length). The purpose of this review is to describe the crucial aspects of genome instability, to outline the ways in which environmental chemicals can affect this cancer hallmark and to identify candidate chemicals for further study. The overall aim is to make scientists aware of the increasing need to unravel the underlying mechanisms via which chemicals at low doses can induce genome instability and thus promote carcinogenesis.
Collapse
Affiliation(s)
- Sabine A S Langie
- Environmental Risk and Health Unit, Flemish Institute for Technological Research (VITO), Boeretang 200, 2400 Mol, Belgium, Health Canada, Environmental Health Sciences and Research Bureau, Environmental Health Centre, Ottawa, Ontario K1A0K9, Canada, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Experimental and Clinical Medicine, University of Firenze, Florence 50134, Italy, Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Navarra, Pamplona 31009, Spain, Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA, Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Department of Chemicals and Radiation, Division of Environmental Medicine, Norwegian Institute of Public Health, PO Box 4404, N-0403 Oslo, Norway, Hopkins Building, School of Biological Sciences, University of Reading, Reading, Berkshire RG6 6UB, UK, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Human and Environmental Safety Research, Department of Health Sciences, College of North Atlantic, Doha, State of Qatar, Mediterranean Institute of Oncology, 95029 Viagrande, Italy, Laboratory for Cell Genetics, Vrije Universiteit Brussel, Brussels 1050, Belgium, Department of Biomedical Science, Faculty of Medicine and Health Sciences, University Putra, Serdang 43400, Selangor, Malaysia, University of Copenhagen, Department of Public Health, Copenhagen 1353, Denmark, Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy, Medical Phys
| | - Gudrun Koppen
- Environmental Risk and Health Unit, Flemish Institute for Technological Research (VITO), Boeretang 200, 2400 Mol, Belgium, Health Canada, Environmental Health Sciences and Research Bureau, Environmental Health Centre, Ottawa, Ontario K1A0K9, Canada, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Experimental and Clinical Medicine, University of Firenze, Florence 50134, Italy, Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Navarra, Pamplona 31009, Spain, Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA, Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Department of Chemicals and Radiation, Division of Environmental Medicine, Norwegian Institute of Public Health, PO Box 4404, N-0403 Oslo, Norway, Hopkins Building, School of Biological Sciences, University of Reading, Reading, Berkshire RG6 6UB, UK, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Human and Environmental Safety Research, Department of Health Sciences, College of North Atlantic, Doha, State of Qatar, Mediterranean Institute of Oncology, 95029 Viagrande, Italy, Laboratory for Cell Genetics, Vrije Universiteit Brussel, Brussels 1050, Belgium, Department of Biomedical Science, Faculty of Medicine and Health Sciences, University Putra, Serdang 43400, Selangor, Malaysia, University of Copenhagen, Department of Public Health, Copenhagen 1353, Denmark, Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy, Medical Phys
| | - Daniel Desaulniers
- Health Canada, Environmental Health Sciences and Research Bureau, Environmental Health Centre, Ottawa, Ontario K1A0K9, Canada
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | | | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, Florence 50134, Italy
| | - Amaya Azqueta
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Navarra, Pamplona 31009, Spain
| | - William H Bisson
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA
| | - Dustin G Brown
- Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Gunnar Brunborg
- Department of Chemicals and Radiation, Division of Environmental Medicine, Norwegian Institute of Public Health, PO Box 4404, N-0403 Oslo, Norway
| | - Amelia K Charles
- Hopkins Building, School of Biological Sciences, University of Reading, Reading, Berkshire RG6 6UB, UK
| | - Tao Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA
| | - Annamaria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Firouz Darroudi
- Human and Environmental Safety Research, Department of Health Sciences, College of North Atlantic, Doha, State of Qatar
| | - Stefano Forte
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Laetitia Gonzalez
- Laboratory for Cell Genetics, Vrije Universiteit Brussel, Brussels 1050, Belgium
| | - Roslida A Hamid
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, University Putra, Serdang 43400, Selangor, Malaysia
| | - Lisbeth E Knudsen
- University of Copenhagen, Department of Public Health, Copenhagen 1353, Denmark
| | - Luc Leyns
- Laboratory for Cell Genetics, Vrije Universiteit Brussel, Brussels 1050, Belgium
| | | | - Lorenzo Memeo
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Chiara Mondello
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - Carmel Mothersill
- Medical Physics & Applied Radiation Sciences, McMaster University, Hamilton, Ontario L8S4L8, Canada
| | - Ann-Karin Olsen
- Department of Chemicals and Radiation, Division of Environmental Medicine, Norwegian Institute of Public Health, PO Box 4404, N-0403 Oslo, Norway
| | - Sofia Pavanello
- Department of Cardiac, Thoracic and Vascular Sciences, Unit of Occupational Medicine, University of Padova, Padova 35128, Italy
| | - Jayadev Raju
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Emilio Rojas
- Departamento de Medicina Genomica y Toxicologia Ambiental, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de México, México CP 04510, México
| | - Rabindra Roy
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences/Food Science and Human Nutrition, College of Veterinary Medicine and Biomedical Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Patricia Ostrosky-Wegman
- Departamento de Medicina Genomica y Toxicologia Ambiental, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de México, México CP 04510, México
| | - Hosni K Salem
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - A Ivana Scovassi
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - Neetu Singh
- Centre for Advanced Research, King George's Medical University, Chowk, Lucknow 226003, Uttar Pradesh, India
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Frederik J Van Schooten
- Department of Toxicology, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University, 6200MD, PO Box 61, Maastricht, The Netherlands
| | - Mahara Valverde
- Departamento de Medicina Genomica y Toxicologia Ambiental, Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de México, México CP 04510, México
| | - Jordan Woodrick
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Luoping Zhang
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA 94720-7360, USA
| | - Nik van Larebeke
- Laboratory for Analytical and Environmental Chemistry, Vrije Universiteit Brussel, Brussels 1050, Belgium, Study Centre for Carcinogenesis and Primary Prevention of Cancer, Ghent University, Ghent 9000, Belgium
| | | | | |
Collapse
|
21
|
Engström W, Darbre P, Eriksson S, Gulliver L, Hultman T, Karamouzis MV, Klaunig JE, Mehta R, Moorwood K, Sanderson T, Sone H, Vadgama P, Wagemaker G, Ward A, Singh N, Al-Mulla F, Al-Temaimi R, Amedei A, Colacci AM, Vaccari M, Mondello C, Scovassi AI, Raju J, Hamid RA, Memeo L, Forte S, Roy R, Woodrick J, Salem HK, Ryan EP, Brown DG, Bisson WH. The potential for chemical mixtures from the environment to enable the cancer hallmark of sustained proliferative signalling. Carcinogenesis 2015; 36 Suppl 1:S38-60. [PMID: 26106143 DOI: 10.1093/carcin/bgv030] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The aim of this work is to review current knowledge relating the established cancer hallmark, sustained cell proliferation to the existence of chemicals present as low dose mixtures in the environment. Normal cell proliferation is under tight control, i.e. cells respond to a signal to proliferate, and although most cells continue to proliferate into adult life, the multiplication ceases once the stimulatory signal disappears or if the cells are exposed to growth inhibitory signals. Under such circumstances, normal cells remain quiescent until they are stimulated to resume further proliferation. In contrast, tumour cells are unable to halt proliferation, either when subjected to growth inhibitory signals or in the absence of growth stimulatory signals. Environmental chemicals with carcinogenic potential may cause sustained cell proliferation by interfering with some cell proliferation control mechanisms committing cells to an indefinite proliferative span.
Collapse
Affiliation(s)
- Wilhelm Engström
- Department of Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Swedish University of Agricultural Sciences, PO Box 7028, 75007 Uppsala, Sweden,
| | - Philippa Darbre
- School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6UB, UK
| | - Staffan Eriksson
- Department of Biochemistry, Faculty of Veterinary Medicine, Swedish University of Agricultural Sciences, Box 575, 75123 Uppsala, Sweden
| | - Linda Gulliver
- Faculty of Medicine, University of Otago, PO Box 913, Dunedin 9050, New Zealand
| | - Tove Hultman
- Department of Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Swedish University of Agricultural Sciences, PO Box 7028, 75007 Uppsala, Sweden, School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6UB, UK
| | - Michalis V Karamouzis
- Department of Biological Chemistry Medical School, Institute of Molecular Medicine and Biomedical Research, University of Athens, Marasli 3, Kolonaki, Athens 10676, Greece
| | - James E Klaunig
- Department of Environmental Health, School of Public Health, Indiana University Bloomington , 1025 E. 7th Street, Suite 111, Bloomington, IN 47405, USA
| | - Rekha Mehta
- Regulatory Toxicology Research Division, Bureau of Chemical Safety, Food Directorate, HPFB, Health Canada, 251 Sir F.G. Banting Driveway, AL # 2202C, Tunney's Pasture, Ottawa, Ontario K1A 0K9, Canada
| | - Kim Moorwood
- Department of Biochemistry and Biology, University of Bath , Claverton Down, Bath BA2 7AY, UK
| | - Thomas Sanderson
- INRS-Institut Armand-Frappier, 531 boulevard des Prairies, Laval, Quebec H7V 1B7, Canada
| | - Hideko Sone
- Environmental Exposure Research Section, Center for Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, Ibraki 3058506, Japan
| | - Pankaj Vadgama
- IRC in Biomedical Materials, School of Engineering & Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Gerard Wagemaker
- Center for Stem Cell Research and Development, Hacettepe University, Ankara 06100, Turkey
| | - Andrew Ward
- Department of Biochemistry and Biology, University of Bath , Claverton Down, Bath BA2 7AY, UK
| | - Neetu Singh
- Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | | | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze 50134, Italy
| | - Anna Maria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Chiara Mondello
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - A Ivana Scovassi
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - Jayadev Raju
- Regulatoty Toxicology Research Division, Bureau of Chemical Safety, Food Directorate, HPFB, Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Roslida A Hamid
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Stefano Forte
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Rabindra Roy
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Jordan Woodrick
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Hosni K Salem
- Urology Dept. kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Sciences, Colorado State University//Colorado School of Public Health, Fort Collins CO 80523-1680, USA and
| | - Dustin G Brown
- Department of Environmental and Radiological Sciences, Colorado State University//Colorado School of Public Health, Fort Collins CO 80523-1680, USA and
| | - William H Bisson
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
22
|
Thompson PA, Khatami M, Baglole CJ, Sun J, Harris SA, Moon EY, Al-Mulla F, Al-Temaimi R, Brown DG, Colacci A, Mondello C, Raju J, Ryan EP, Woodrick J, Scovassi AI, Singh N, Vaccari M, Roy R, Forte S, Memeo L, Salem HK, Amedei A, Hamid RA, Lowe L, Guarnieri T, Bisson WH. Environmental immune disruptors, inflammation and cancer risk. Carcinogenesis 2015; 36 Suppl 1:S232-53. [PMID: 26106141 DOI: 10.1093/carcin/bgv038] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
An emerging area in environmental toxicology is the role that chemicals and chemical mixtures have on the cells of the human immune system. This is an important area of research that has been most widely pursued in relation to autoimmune diseases and allergy/asthma as opposed to cancer causation. This is despite the well-recognized role that innate and adaptive immunity play as essential factors in tumorigenesis. Here, we review the role that the innate immune cells of inflammatory responses play in tumorigenesis. Focus is placed on the molecules and pathways that have been mechanistically linked with tumor-associated inflammation. Within the context of chemically induced disturbances in immune function as co-factors in carcinogenesis, the evidence linking environmental toxicant exposures with perturbation in the balance between pro- and anti-inflammatory responses is reviewed. Reported effects of bisphenol A, atrazine, phthalates and other common toxicants on molecular and cellular targets involved in tumor-associated inflammation (e.g. cyclooxygenase/prostaglandin E2, nuclear factor kappa B, nitric oxide synthesis, cytokines and chemokines) are presented as example chemically mediated target molecule perturbations relevant to cancer. Commentary on areas of additional research including the need for innovation and integration of systems biology approaches to the study of environmental exposures and cancer causation are presented.
Collapse
Affiliation(s)
- Patricia A Thompson
- Department of Pathology, Stony Brook Medical School, Stony Brook, NY 11794, USA, Inflammation and Cancer Research, National Cancer Institute (NCI) (Retired), NIH, Bethesda, MD 20817, USA, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada, Department of Biochemistry, Rush University, Chicago, IL 60612, USA, Prevention and Cancer Control, Cancer Care Ontario, 620 University Avenue, Toronto, Ontario M5G 2L3, Canada, Department of Bioscience and Biotechnology, Sejong University, Seoul 143-747, Republic of South Korea, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Environmental and Radiological Health Sciences, Colorado State University, Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy, The Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy, Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA, Advanced Molecular Science Research Centre, King George's Medical University, Lucknow, Uttar Pradesh 226003, India, Mediterranean Institute of Oncology, 95029 Viagrande, Italy, Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt, Department of Experimental and Clinical Medicine, University of Firenze, 50134 Florence, Italy, Faculty of Medicine and Health Sciences, Universiti Putra, Malaysia, Serdang, Selangor 43400, Malaysia, Getting to Know Cancer, Room 229A, 36 Arthur St, Truro, Nova Scotia B2N 1X5, Canada Department of Biology, Geology and Environmental Sciences, Alma Mater Studiorum Università di Bologna, Via Francesco Selmi, 3, 40126 Bologna, Italy Center for Appl
| | - Mahin Khatami
- Inflammation and Cancer Research, National Cancer Institute (NCI) (Retired), NIH, Bethesda, MD 20817, USA
| | - Carolyn J Baglole
- Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada
| | - Jun Sun
- Department of Biochemistry, Rush University, Chicago, IL 60612, USA
| | - Shelley A Harris
- Prevention and Cancer Control, Cancer Care Ontario, 620 University Avenue, Toronto, Ontario M5G 2L3, Canada
| | - Eun-Yi Moon
- Department of Bioscience and Biotechnology, Sejong University, Seoul 143-747, Republic of South Korea
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | | | - Dustin G Brown
- Department of Environmental and Radiological Health Sciences, Colorado State University, Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Annamaria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | - Chiara Mondello
- The Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy
| | - Jayadev Raju
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences, Colorado State University, Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Jordan Woodrick
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - A Ivana Scovassi
- The Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy
| | - Neetu Singh
- Advanced Molecular Science Research Centre, King George's Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | - Rabindra Roy
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - Stefano Forte
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Hosni K Salem
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, 50134 Florence, Italy
| | - Roslida A Hamid
- Faculty of Medicine and Health Sciences, Universiti Putra, Malaysia, Serdang, Selangor 43400, Malaysia
| | - Leroy Lowe
- Getting to Know Cancer, Room 229A, 36 Arthur St, Truro, Nova Scotia B2N 1X5, Canada
| | - Tiziana Guarnieri
- Department of Biology, Geology and Environmental Sciences, Alma Mater Studiorum Università di Bologna, Via Francesco Selmi, 3, 40126 Bologna, Italy Center for Applied Biomedical Research, S. Orsola-Malpighi University Hospital, Via Massarenti, 9, 40126 Bologna, Italy, National Institute of Biostructures and Biosystems, Viale Medaglie d' Oro, 305, 00136 Roma, Italy and
| | - William H Bisson
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, Oregon 97331, USA
| |
Collapse
|
23
|
Nahta R, Al-Mulla F, Al-Temaimi R, Amedei A, Andrade-Vieira R, Bay SN, Brown DG, Calaf GM, Castellino RC, Cohen-Solal KA, Colacci A, Cruickshanks N, Dent P, Di Fiore R, Forte S, Goldberg GS, Hamid RA, Krishnan H, Laird DW, Lasfar A, Marignani PA, Memeo L, Mondello C, Naus CC, Ponce-Cusi R, Raju J, Roy D, Roy R, Ryan EP, Salem HK, Scovassi AI, Singh N, Vaccari M, Vento R, Vondráček J, Wade M, Woodrick J, Bisson WH. Mechanisms of environmental chemicals that enable the cancer hallmark of evasion of growth suppression. Carcinogenesis 2015; 36 Suppl 1:S2-18. [PMID: 26106139 DOI: 10.1093/carcin/bgv028] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
As part of the Halifax Project, this review brings attention to the potential effects of environmental chemicals on important molecular and cellular regulators of the cancer hallmark of evading growth suppression. Specifically, we review the mechanisms by which cancer cells escape the growth-inhibitory signals of p53, retinoblastoma protein, transforming growth factor-beta, gap junctions and contact inhibition. We discuss the effects of selected environmental chemicals on these mechanisms of growth inhibition and cross-reference the effects of these chemicals in other classical cancer hallmarks.
Collapse
Affiliation(s)
- Rita Nahta
- Departments of Pharmacology and Hematology & Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA 30322, USA, Department of Pathology, Kuwait University, Safat 13110, Kuwait, Department of Experimental and Clinical Medicine, University of Firenze, 50134 Florence, Italy, Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada, Program in Genetics and Molecular Biology, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA 30322, USA, Department of Environmental and Radiological Health Sciences/Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Center for Radiological Research, Columbia University Medical Center, New York, NY 10032, USA, Instituto de Alta Investigacion, Universidad de Tarapaca, Arica 8097877, Chile, Division of Hematology and Oncology, Department of Pediatrics, Children's Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA, Department of Medicine/Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901-1914, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy, Departments of Neurosurgery and Biochemistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 980033, USA, Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies, Polyclinic Plexus, University of Palermo, 90127 Palermo, Italy, Mediterranean Institute of Oncology, 95029 Viagrande, Italy, Graduate School of Biomedical Sciences and Department of Molecular Biology, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084-1501, USA, Department of Biomedical Science, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia, Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontari
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | | | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, 50134 Florence, Italy
| | - Rafaela Andrade-Vieira
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Sarah N Bay
- Program in Genetics and Molecular Biology, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA 30322, USA
| | - Dustin G Brown
- Department of Environmental and Radiological Health Sciences/Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Gloria M Calaf
- Center for Radiological Research, Columbia University Medical Center, New York, NY 10032, USA, Instituto de Alta Investigacion, Universidad de Tarapaca, Arica 8097877, Chile
| | - Robert C Castellino
- Division of Hematology and Oncology, Department of Pediatrics, Children's Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA
| | - Karine A Cohen-Solal
- Department of Medicine/Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901-1914, USA
| | - Annamaria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Nichola Cruickshanks
- Departments of Neurosurgery and Biochemistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 980033, USA
| | - Paul Dent
- Departments of Neurosurgery and Biochemistry and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 980033, USA
| | - Riccardo Di Fiore
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies, Polyclinic Plexus, University of Palermo, 90127 Palermo, Italy
| | - Stefano Forte
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Gary S Goldberg
- Graduate School of Biomedical Sciences and Department of Molecular Biology, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084-1501, USA
| | - Roslida A Hamid
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor 43400, Malaysia
| | - Harini Krishnan
- Graduate School of Biomedical Sciences and Department of Molecular Biology, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084-1501, USA
| | - Dale W Laird
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Ahmed Lasfar
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, NJ 60503, USA
| | - Paola A Marignani
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Chiara Mondello
- Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy
| | - Christian C Naus
- Department of Cellular & Physiological Sciences, Life Sciences Institute, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Richard Ponce-Cusi
- Instituto de Alta Investigacion, Universidad de Tarapaca, Arica 8097877, Chile
| | - Jayadev Raju
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Debasish Roy
- Department of Natural Science, The City University of New York at Hostos Campus, Bronx, NY 10451, USA
| | - Rabindra Roy
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences/Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Hosni K Salem
- Urology Dept., kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - A Ivana Scovassi
- Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy
| | - Neetu Singh
- Advanced Molecular Science Research Centre, King George's Medical University, Lucknow, UP 226003, India
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Renza Vento
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies, Polyclinic Plexus, University of Palermo, 90127 Palermo, Italy, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Jan Vondráček
- Department of Cytokinetics, Institute of Biophysics AS CR, Brno 612 65, Czech Republic
| | - Mark Wade
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia (IIT), Milan 16163, Italy and
| | - Jordan Woodrick
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - William H Bisson
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
24
|
Casey SC, Vaccari M, Al-Mulla F, Al-Temaimi R, Amedei A, Barcellos-Hoff MH, Brown DG, Chapellier M, Christopher J, Curran CS, Forte S, Hamid RA, Heneberg P, Koch DC, Krishnakumar PK, Laconi E, Maguer-Satta V, Marongiu F, Memeo L, Mondello C, Raju J, Roman J, Roy R, Ryan EP, Ryeom S, Salem HK, Scovassi AI, Singh N, Soucek L, Vermeulen L, Whitfield JR, Woodrick J, Colacci A, Bisson WH, Felsher DW. The effect of environmental chemicals on the tumor microenvironment. Carcinogenesis 2015; 36 Suppl 1:S160-83. [PMID: 26106136 DOI: 10.1093/carcin/bgv035] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Potentially carcinogenic compounds may cause cancer through direct DNA damage or through indirect cellular or physiological effects. To study possible carcinogens, the fields of endocrinology, genetics, epigenetics, medicine, environmental health, toxicology, pharmacology and oncology must be considered. Disruptive chemicals may also contribute to multiple stages of tumor development through effects on the tumor microenvironment. In turn, the tumor microenvironment consists of a complex interaction among blood vessels that feed the tumor, the extracellular matrix that provides structural and biochemical support, signaling molecules that send messages and soluble factors such as cytokines. The tumor microenvironment also consists of many host cellular effectors including multipotent stromal cells/mesenchymal stem cells, fibroblasts, endothelial cell precursors, antigen-presenting cells, lymphocytes and innate immune cells. Carcinogens can influence the tumor microenvironment through effects on epithelial cells, the most common origin of cancer, as well as on stromal cells, extracellular matrix components and immune cells. Here, we review how environmental exposures can perturb the tumor microenvironment. We suggest a role for disrupting chemicals such as nickel chloride, Bisphenol A, butyltins, methylmercury and paraquat as well as more traditional carcinogens, such as radiation, and pharmaceuticals, such as diabetes medications, in the disruption of the tumor microenvironment. Further studies interrogating the role of chemicals and their mixtures in dose-dependent effects on the tumor microenvironment could have important general mechanistic implications for the etiology and prevention of tumorigenesis.
Collapse
Affiliation(s)
- Stephanie C Casey
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA 94305, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy, Department of Pathology, Kuwait University, 13110 Safat, Kuwait, Department of Experimental and Clinical Medicine, University of Firenze, 50134 Florence, Italy, Department of Radiation Oncology, NYU School of Medicine, New York, NY 10016, USA, Department of Environmental and Radiological Health Sciences, Colorado State University/ Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Centre De Recherche En Cancerologie De Lyon, U1052-UMR5286, Université de Lyon, 69007 Lyon, France, Cancer Research UK, Cambridge Institute, University of Cambridge, Robinson Way, CB2 0RE Cambridge, UK, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA, Mediterranean Institute of Oncology, 95029 Viagrande, Italy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400 Selangor, Malaysia, Charles University in Prague, Third Faculty of Medicine, 100 00 Prague 10, Czech Republic, Center for Environment and Water, Research Institute, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia, Department of Science and Biomedical Technology, University of Cagliari, 09124 Cagliari, Italy, Pathology Unit, Mediterranean Institute of Oncology, 95029 Viagrande, Italy, Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy, Regulatory Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Department of Medicine, University of Louisville, Louisville, KY 40202, USA, Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA, University of Pennsylvania School of Medicine
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, 13110 Safat, Kuwait
| | | | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, 50134 Florence, Italy
| | | | - Dustin G Brown
- Department of Environmental and Radiological Health Sciences, Colorado State University/ Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Marion Chapellier
- Centre De Recherche En Cancerologie De Lyon, U1052-UMR5286, Université de Lyon, 69007 Lyon, France
| | - Joseph Christopher
- Cancer Research UK, Cambridge Institute, University of Cambridge, Robinson Way, CB2 0RE Cambridge, UK
| | - Colleen S Curran
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Stefano Forte
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Roslida A Hamid
- Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400 Selangor, Malaysia
| | - Petr Heneberg
- Charles University in Prague, Third Faculty of Medicine, 100 00 Prague 10, Czech Republic
| | - Daniel C Koch
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA 94305, USA, Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy, Department of Pathology, Kuwait University, 13110 Safat, Kuwait, Department of Experimental and Clinical Medicine, University of Firenze, 50134 Florence, Italy, Department of Radiation Oncology, NYU School of Medicine, New York, NY 10016, USA, Department of Environmental and Radiological Health Sciences, Colorado State University/ Colorado School of Public Health, Fort Collins, CO 80523-1680, USA, Centre De Recherche En Cancerologie De Lyon, U1052-UMR5286, Université de Lyon, 69007 Lyon, France, Cancer Research UK, Cambridge Institute, University of Cambridge, Robinson Way, CB2 0RE Cambridge, UK, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA, Mediterranean Institute of Oncology, 95029 Viagrande, Italy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400 Selangor, Malaysia, Charles University in Prague, Third Faculty of Medicine, 100 00 Prague 10, Czech Republic, Center for Environment and Water, Research Institute, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia, Department of Science and Biomedical Technology, University of Cagliari, 09124 Cagliari, Italy, Pathology Unit, Mediterranean Institute of Oncology, 95029 Viagrande, Italy, Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy, Regulatory Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada, Department of Medicine, University of Louisville, Louisville, KY 40202, USA, Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA, University of Pennsylvania School of Medicine
| | - P K Krishnakumar
- Center for Environment and Water, Research Institute, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia
| | - Ezio Laconi
- Department of Science and Biomedical Technology, University of Cagliari, 09124 Cagliari, Italy
| | - Veronique Maguer-Satta
- Centre De Recherche En Cancerologie De Lyon, U1052-UMR5286, Université de Lyon, 69007 Lyon, France
| | - Fabio Marongiu
- Department of Science and Biomedical Technology, University of Cagliari, 09124 Cagliari, Italy
| | - Lorenzo Memeo
- Pathology Unit, Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Chiara Mondello
- Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy
| | - Jayadev Raju
- Regulatory Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Jesse Roman
- Department of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Rabindra Roy
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences, Colorado State University/ Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Sandra Ryeom
- University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Hosni K Salem
- Urology Department, Kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 11562, Egypt
| | - A Ivana Scovassi
- Institute of Molecular Genetics, National Research Council, 27100 Pavia, Italy
| | - Neetu Singh
- Centre for Advanced Research, King George's Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India
| | - Laura Soucek
- Vall d'Hebron Institute of Oncology (VHIO) and Institució Catalana de Recerca i Estudis Avançats (ICREA), 08035 Barcelona, Spain
| | - Louis Vermeulen
- Center for Experimental Molecular Medicine (CEMM), Academic Medical Center (AMC), Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Jonathan R Whitfield
- Vall d'Hebron Institute of Oncology (VHIO) and Institució Catalana de Recerca i Estudis Avançats (ICREA), 08035 Barcelona, Spain
| | - Jordan Woodrick
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20057, USA
| | - Annamaria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | - William H Bisson
- Department of Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA, and
| | - Dean W Felsher
- Division of Oncology, Departments of Medicine and Pathology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
25
|
Kass L, Durando M, Altamirano GA, Manfroni-Ghibaudo GE, Luque EH, Muñoz-de-Toro M. Prenatal Bisphenol A exposure delays the development of the male rat mammary gland. Reprod Toxicol 2015; 54:37-46. [DOI: 10.1016/j.reprotox.2014.02.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 12/30/2013] [Accepted: 02/13/2014] [Indexed: 12/22/2022]
|
26
|
Kravchenko J, Corsini E, Williams MA, Decker W, Manjili MH, Otsuki T, Singh N, Al-Mulla F, Al-Temaimi R, Amedei A, Colacci AM, Vaccari M, Mondello C, Scovassi AI, Raju J, Hamid RA, Memeo L, Forte S, Roy R, Woodrick J, Salem HK, Ryan EP, Brown DG, Bisson WH, Lowe L, Lyerly HK. Chemical compounds from anthropogenic environment and immune evasion mechanisms: potential interactions. Carcinogenesis 2015; 36 Suppl 1:S111-27. [PMID: 26002081 DOI: 10.1093/carcin/bgv033] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 01/19/2015] [Indexed: 02/07/2023] Open
Abstract
An increasing number of studies suggest an important role of host immunity as a barrier to tumor formation and progression. Complex mechanisms and multiple pathways are involved in evading innate and adaptive immune responses, with a broad spectrum of chemicals displaying the potential to adversely influence immunosurveillance. The evaluation of the cumulative effects of low-dose exposures from the occupational and natural environment, especially if multiple chemicals target the same gene(s) or pathway(s), is a challenge. We reviewed common environmental chemicals and discussed their potential effects on immunosurveillance. Our overarching objective was to review related signaling pathways influencing immune surveillance such as the pathways involving PI3K/Akt, chemokines, TGF-β, FAK, IGF-1, HIF-1α, IL-6, IL-1α, CTLA-4 and PD-1/PDL-1 could individually or collectively impact immunosurveillance. A number of chemicals that are common in the anthropogenic environment such as fungicides (maneb, fluoxastrobin and pyroclostrobin), herbicides (atrazine), insecticides (pyridaben and azamethiphos), the components of personal care products (triclosan and bisphenol A) and diethylhexylphthalate with pathways critical to tumor immunosurveillance. At this time, these chemicals are not recognized as human carcinogens; however, it is known that they these chemicalscan simultaneously persist in the environment and appear to have some potential interfere with the host immune response, therefore potentially contributing to promotion interacting with of immune evasion mechanisms, and promoting subsequent tumor growth and progression.
Collapse
Affiliation(s)
- Julia Kravchenko
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA;
| | - Emanuela Corsini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, School of Pharmacy, Università degli Studi di Milano, 20133 Milan, Italy
| | - Marc A Williams
- MEDCOM Army Institute of Public Health, Toxicology Portfolio - Health Effects Research Program, Aberdeen Proving Ground, Edgewood, Baltimore, MD 21010, USA
| | - William Decker
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Masoud H Manjili
- Department of Microbiology and Immunology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Takemi Otsuki
- Department of Hygiene, Kawasaki Medical School, Kurashiki 701-0192, Japan
| | - Neetu Singh
- Advanced Molecular Science Research Centre, King George's Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Faha Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | | | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze 50134, Italy
| | - Anna Maria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, 40126 Bologna, Italy
| | - Chiara Mondello
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - A Ivana Scovassi
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - Jayadev Raju
- Toxicology Research Division, Bureau of Chemical Safety, Food Directorate, HPFB, Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Roslida A Hamid
- Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor 43400, Malaysia
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Stefano Forte
- Mediterranean Institute of Oncology, 95029 Viagrande, Italy
| | - Rabindra Roy
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Jordan Woodrick
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Hosni K Salem
- Urology Department, Kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences, Colorado State University/ Colorado School of Public Health, Fort Collins, CO, 80523-1680, USA
| | - Dustin G Brown
- Department of Environmental and Radiological Health Sciences, Colorado State University/ Colorado School of Public Health, Fort Collins, CO, 80523-1680, USA
| | - William H Bisson
- Environmental and Molecular Toxicology, Environmental Health Sciences Center, Oregon State University, Corvallis, OR 97331, USA,
| | - Leroy Lowe
- Getting to Know Cancer, Nova Scotia, Canada and
| | - H Kim Lyerly
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA; Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
27
|
Chen J, Zhang X, Wang Y, Ye Y, Huang Z. Formononetin promotes proliferation that involves a feedback loop of microRNA-375 and estrogen receptor alpha in estrogen receptor-positive cells. Mol Carcinog 2015; 55:312-9. [PMID: 25663261 DOI: 10.1002/mc.22282] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 11/30/2014] [Accepted: 12/18/2014] [Indexed: 12/29/2022]
Abstract
Formononetin is an O-methylated isoflavone that is isolated from the root of Astragalus membranaceus, and it has antitumorigenic effects. Our previous studies found that formononetin triggered growth-inhibitory and apoptotic activities in MCF-7 breast cancer cells. To further investigate the potential effect of formononetin in promoting cell proliferation in estrogen receptor (ER)-positive cells, we used in vivo and in vitro studies to elucidate the possible mechanism. ERα-positive cells (HUVEC, MCF-7) were treated with formononetin. The CCK8 assay, Hoechst 33258, and flow cytometry were used to assess cell proliferation and apoptosis. mRNA levels of ERα, Bcl-2, and miR-375 were quantified using real-time polymerase chain reaction. ERα, p-Akt, and Bcl-2 expression was determined using Western blot. Compared with the control, low formononetin concentrations (2-6 μM) stimulated ERα-positive cell proliferation (HUVEC, MCF-7). The more sensitive HUVEC cells were used to study the relevant signaling pathway. After treatment with formononetin, ERα, miR-375, p-Akt, and Bcl-2 expression was significantly upregulated. The proliferative effect of formononetin was also blocked by a miR-375 inhibitor or raloxifene pretreatment. Additionally, in the in vivo studies, uterine weight in ovariectomized mice treated with formononetin increased significantly, but the weight dramatically decreased with raloxifene or miR-375 inhibitor pretreatment before formononetin. This study demonstrated that formononetin promoted ERα-positive cell proliferation through miR-375 activation and this mechanism is possibly involving in a miR-375 and ERα feedback loop.
Collapse
Affiliation(s)
- Jian Chen
- School of Basic Medical Sciences, Guilin Medical University, Guilin, China
| | - Xing Zhang
- School of Basic Medical Sciences, Guilin Medical University, Guilin, China
| | - Yong Wang
- School of Basic Medical Sciences, Guilin Medical University, Guilin, China
| | - Yu Ye
- Department of Emergency, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhaoquan Huang
- Department of Pathology, Guilin Medical University, Guilin, China
| |
Collapse
|
28
|
Liu J, Jin X, Zhao N, Ye X, Ying C. Bisphenol A promotes X-linked inhibitor of apoptosis protein-dependent angiogenesis via G protein-coupled estrogen receptor pathway. J Appl Toxicol 2015; 35:1309-17. [PMID: 25663485 DOI: 10.1002/jat.3112] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 11/18/2014] [Accepted: 12/04/2014] [Indexed: 02/06/2023]
Abstract
Bisphenol A (BPA), one of the high-volume chemicals worldwide, has a core structure resembling that of natural estradiol. Recent evidence has demonstrated that exposure to BPA has a relationship with the risk of cancer. The objective of our study is to investigate the mechanisms underlying the pro-angiogenic effects of BPA. We demonstrated that BPA markedly induces endothelial cell proliferation, migration and tube formation by activating endothelial nitric oxide synthase. BPA-induced nitric oxide generation appeared to be associated with the X-linked inhibitor of apoptosis protein (XIAP), which competes with endothelial nitric oxide synthase for caveolin-1. BPA was shown to exert its pro-angiogenic effect by upregulating XIAP expression via G protein-coupled estrogen receptor (ER) activation but not via ERα or ERβ. Our data suggest that 100 nM BPA promote angiogenesis in a G protein-coupled ER-dependent genomic pathway, and provide a novel insight into the potential role of XIAP in mediating the pro-angiogenic effects of BPA in endothelial cells.
Collapse
Affiliation(s)
- Jian Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.,Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Xin Jin
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Nana Zhao
- School of Environmental Science and Public Health, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Xiaolei Ye
- School of Environmental Science and Public Health, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Chenjiang Ying
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.,MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
29
|
Milesi MM, Alarcón R, Ramos JG, Muñoz-de-Toro M, Luque EH, Varayoud J. Neonatal exposure to low doses of endosulfan induces implantation failure and disrupts uterine functional differentiation at the pre-implantation period in rats. Mol Cell Endocrinol 2015; 401:248-59. [PMID: 25486513 DOI: 10.1016/j.mce.2014.11.028] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 11/26/2014] [Accepted: 11/28/2014] [Indexed: 01/12/2023]
Abstract
We investigated whether neonatal exposure to low doses of endosulfan affects fertility and uterine functional differentiation at pre-implantation in rats. Newborn female rats received the vehicle, 0.2 µg/kg/d of diethylstilbestrol (DES), 6 µg/kg/d of endosulfan (Endo6) or 600 µg/kg/d of endosulfan (Endo600) on postnatal days (PND) 1, 3, 5, and 7. On PND90, the rats were mated to evaluate their reproductive performance on gestational day (GD) 19 and their ovarian steroid serum levels, endometrial proliferation and implantation-associated proteins on GD5. DES and endosulfan decreased the pregnancy rate and the number of implantation sites. On GD5, DES and endosulfan did not change the serum levels of 17β-estradiol (E2) and progesterone (P); the endometrial proliferation decreased, which was associated with silencing of Hoxa10 in the Endo600-treated rats. Both doses of endosulfan increased the progesterone receptor (PR) expression, whereas the higher dose led additionally to an increase in estrogen receptor alpha (ERα). In the Endo600-treated rats, the down-regulation of Hoxa10 was associated with a deregulation of the steroid receptor coregulators. Alterations in endometrial proliferation and the endocrine pathway of Hoxa10/steroid receptors/coregulators might be the mechanism of endosulfan-induced implantation failure.
Collapse
Affiliation(s)
- María M Milesi
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - Ramiro Alarcón
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - Jorge G Ramos
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - Mónica Muñoz-de-Toro
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - Enrique H Luque
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - Jorgelina Varayoud
- Instituto de Salud y Ambiente del Litoral (ISAL), Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina.
| |
Collapse
|
30
|
Vandenberg LN, Ehrlich S, Belcher SM, Ben-Jonathan N, Dolinoy DC, Hugo ER, Hunt PA, Newbold RR, Rubin BS, Saili KS, Soto AM, Wang HS, vom Saal FS. Low dose effects of bisphenol A. ACTA ACUST UNITED AC 2014. [DOI: 10.4161/endo.26490] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
31
|
Trabert B, Falk RT, Figueroa JD, Graubard BI, Garcia-Closas M, Lissowska J, Peplonska B, Fox SD, Brinton LA. Urinary bisphenol A-glucuronide and postmenopausal breast cancer in Poland. Cancer Causes Control 2014; 25:1587-93. [PMID: 25189422 DOI: 10.1007/s10552-014-0461-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 08/16/2014] [Indexed: 11/30/2022]
Abstract
PURPOSE Concerns regarding a possible link between bisphenol A (BPA) and breast cancer have been mounting, but studies in human populations are lacking. We evaluated the association between the major urinary BPA metabolite [BPA-glucuronide (BPA-G)] and postmenopausal breast cancer risk in a large population-based case-control study conducted in two cities in Poland (2000-2003); we further explored the association of BPA-G levels with known postmenopausal breast cancer risk factors in our control population. METHODS We analyzed creatinine-adjusted urinary BPA-G levels among 575 postmenopausal cases matched on age and study site to 575 controls without breast cancer using a recently developed assay. Odds ratios and 95 % confidence intervals were used to estimate the association between urinary BPA-G level and breast cancer using conditional logistic regression. Among controls, geometric mean BPA-G levels were compared across categories of breast cancer risk factors using linear regression models. RESULTS There was no indication that increased BPA-G was associated with postmenopausal breast cancer (p-trend = 0.59). Among controls, mean BPA-G was higher among women reporting extended use of menopausal hormones, a prior screening mammogram, and residence in Warsaw. Other comparisons across strata of postmenopausal breast cancer risk factors were not related to differences in BPA-G. CONCLUSIONS Urinary BPA-G, measured at the time of diagnosis, is not linked to postmenopausal breast cancer.
Collapse
Affiliation(s)
- Britton Trabert
- Division of Cancer Epidemiology and Genetics, Department of Health and Human Services, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Room 7E-228, Bethesda, MD, 20892-9774, USA,
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Durando M, Cocito L, Rodríguez HA, Varayoud J, Ramos JG, Luque EH, Muñoz-de-Toro M. Neonatal expression of amh, sox9 and sf-1 mRNA in Caiman latirostris and effects of in ovo exposure to endocrine disrupting chemicals. Gen Comp Endocrinol 2013; 191:31-8. [PMID: 23747749 DOI: 10.1016/j.ygcen.2013.05.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 02/26/2013] [Accepted: 05/21/2013] [Indexed: 01/07/2023]
Abstract
Caiman latirostris is a reptilian species that exhibits temperature-dependent sex determination (TSD). Male-to-female sex reversal can be achieved after in ovo estrogen/xenoestrogen exposure. This is known as hormone-dependent sex determination (HSD). The amh, sox9 and sf-1 genes are involved in sex determination, sex differentiation, and steroidogenesis. The aims of this study were: (a) to establish the expression patterns of amh, sox9 and sf-1 mRNA in the gonad-adrenal-mesonephros (GAM) complexes of neonatal TSD-male and TSD-female caimans, (b) to compare the expression of these genes between TSD-females and HSD-females (born from E2-exposed eggs incubated at the male-producing temperature) and (c) to evaluate whether in ovo exposure to a low dose of E2 or bisphenol A (BPA) or to a high dose of endosulfan (END) modifies amh, sox9 or sf-1 mRNA expressions in neonatal males. The mRNA expressions of amh, sox9 and sf-1 in GAM complexes from TSD-males and TSD-females and from HSD-females were quantitatively compared by RT-PCR. A sexually dimorphic pattern of amh and sox9 mRNA expression was found, with a higher expression in TSD-males than in TSD-females. sf-1 mRNA did not differ between TSD-males and TSD-females. HSD-females exhibited a higher expression of sox9 than TSD-females. In males, increased mRNA expression of sex-determining genes was observed after in ovo exposure to END. E2 decreased sox9 but increased sf-1 mRNA expression. Changes induced by BPA were evident although not significant. These results provide new insights into the potential mechanisms that lead to the gonadal histo-functional alterations observed in caimans exposed to contaminated environments.
Collapse
Affiliation(s)
- Milena Durando
- Laboratorio de Endocrinología y Tumores Hormonodependientes, School of Biochemistry and Biological Sciences, Universidad Nacional del Litoral, Santa Fe, Argentina
| | | | | | | | | | | | | |
Collapse
|
33
|
Sharan S, Nikhil K, Roy P. Effects of low dose treatment of tributyltin on the regulation of estrogen receptor functions in MCF-7 cells. Toxicol Appl Pharmacol 2013; 269:176-86. [PMID: 23523586 DOI: 10.1016/j.taap.2013.03.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 03/06/2013] [Accepted: 03/08/2013] [Indexed: 01/15/2023]
|
34
|
Matsuda S, Matsuzawa D, Ishii D, Tomizawa H, Sajiki J, Shimizu E. Perinatal exposure to bisphenol A enhances contextual fear memory and affects the serotoninergic system in juvenile female mice. Horm Behav 2013; 63:709-16. [PMID: 23567477 DOI: 10.1016/j.yhbeh.2013.03.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 03/26/2013] [Accepted: 03/29/2013] [Indexed: 02/06/2023]
Abstract
Perinatal exposure to bisphenol A (BPA), an endocrine-disrupting chemical, affects the central nervous system, including effects on emotional responses and neurotransmitter release. In this study, we investigated the effects of BPA (250 ng/kg/day, from gestational day 10 to postnatal day 20) on fear memory and serotonin (5-HT) metabolites in the brain using contextual fear conditioning (FC) and high-performance liquid chromatography (HPLC), respectively, in adult and juvenile mice of both sexes. Furthermore, we studied the effects of BPA on the gene expression of 5-HT metabolite-related enzymes and 5-HT receptors using quantitative real-time RT PCR in the brains of juvenile females. BPA enhanced fear memory and increased serotonin metabolite (5-HIAA) levels and 5-HIAA/5-HT in the hippocampus, the striatum, the midbrain, the pons, and the medulla oblongata of juvenile female mice. In contrast, alterations in those areas were much smaller in adult females and in both juvenile and adult males. Furthermore, BPA induced increases in the expression levels of Tph2, Slc6a4, and Maoa mRNA in the hippocampus of juvenile females, indicating that BPA induces hyper 5-HT turnover in the hippocampus. Our results suggest that perinatal exposure to a low dose of BPA enhances fear memory and the 5-HTergic system in juvenile mice.
Collapse
Affiliation(s)
- Shingo Matsuda
- Department of Cognitive Behavioral Physiology, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chiba 260-8670, Japan.
| | | | | | | | | | | |
Collapse
|
35
|
Abdel-Rahman WM, Moustafa YM, Ahmed BO, Mostafa RM. Endocrine Disruptors and Breast Cancer Risk - Time to Consider the Environment. Asian Pac J Cancer Prev 2012; 13:5937-5946. [DOI: 10.7314/apjcp.2012.13.12.5937] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024] Open
|
36
|
Basini G, Bianchi F, Bussolati S, Baioni L, Ramoni R, Grolli S, Conti V, Bianchi F, Grasselli F. Atrazine disrupts steroidogenesis, VEGF and NO production in swine granulosa cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2012; 85:59-63. [PMID: 22999709 DOI: 10.1016/j.ecoenv.2012.08.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 08/08/2012] [Accepted: 08/29/2012] [Indexed: 06/01/2023]
Abstract
Atrazine is one of the most widely employed herbicides. Due to its environmental persistence, it can be detected in ground and water thus becoming the subject of a serious concern because of its potential endocrine disrupting activity. In particular, several in vitro and in vivo studies point out adverse effects on reproduction. However, these data were mainly collected in the male, while studies on females are lacking. Present work was therefore set up on swine ovarian granulosa cells to investigate the effect of atrazine on steroidogenesis and proliferation. Moreover, since vessel growth is fundamental for reproductive function, we evaluated the herbicide's effect on two of the main angiogenesis signaling molecules, VEGF and NO. Our data show that atrazine markedly interferes with steroidogenesis while it does not modify cell proliferation; in addition, the herbicide has also been found to affect the production of the examined angiogenesis molecules. Collectively, these results indicate for the first time a potential negative effect of atrazine on ovarian functions in the swine species.
Collapse
Affiliation(s)
- Giuseppina Basini
- Dipartimento di Scienze Medico-Veterinarie, Università degli Studi di Parma, Via del Taglio 10, 43126 Parma, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Knudsen TB, Kleinstreuer NC. Disruption of embryonic vascular development in predictive toxicology. ACTA ACUST UNITED AC 2012; 93:312-23. [DOI: 10.1002/bdrc.20223] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
38
|
Proangiogenic effects of environmentally relevant levels of bisphenol A in human primary endothelial cells. Arch Toxicol 2011; 86:465-74. [DOI: 10.1007/s00204-011-0766-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 09/29/2011] [Indexed: 10/15/2022]
|
39
|
Blumberg B, Iguchi T, Odermatt A. Endocrine disrupting chemicals. J Steroid Biochem Mol Biol 2011; 127:1-3. [PMID: 21839836 DOI: 10.1016/j.jsbmb.2011.07.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 07/28/2011] [Indexed: 01/09/2023]
|