1
|
Gao X, Dan Q, Zhang C, Ding R, Gao E, Luo H, Liu W, Lu C. Pentachloronitrobenzene disturbed murine ventricular wall development by inhibiting cardiomyocyte proliferation via Hec1 downregulation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:168917. [PMID: 38030013 DOI: 10.1016/j.scitotenv.2023.168917] [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: 09/21/2023] [Revised: 11/22/2023] [Accepted: 11/25/2023] [Indexed: 12/01/2023]
Abstract
Exposure to the organochlorine fungicide pentachloronitrobenzene (PCNB) causes developmental abnormalities, including cardiac malformation. However, the molecular mechanism of PCNB cardiotoxicity remains elusive. We found that oral administration of PCNB to pregnant mice induced a hypoplastic wall with significant thinning of the compact myocardium in the developing hearts. PCNB significantly downregulates the expression of Hec1, a member of the NDC80 kinetochore complex, resulting in aberrant spindles, chromosome missegregation and an arrest in cardiomyocyte proliferation. Cardiac-specific ablation of Hec1 sharply inhibits cardiomyocyte proliferation, leading to thinning of the compact myocardium and embryonic lethality. Mechanistically, we found that activating transcription factor 3 (ATF3) transactivates Hec1 expression. Either HEC1 or ATF3 overexpression significantly rescues mitotic defects and restore the decreased proliferative ability of cardiomyocytes caused by PCNB exposure. Our findings highlight that maternal PCNB exposure disrupts embryonic cardiac function by inhibiting cardiomyocyte proliferation and interfering with ventricular wall development, partially attributed to the downregulation of the Atf3-Hec1 axis.
Collapse
Affiliation(s)
- Xiaobo Gao
- Department of Genetics, National Research Institute for Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Qinghua Dan
- Department of Genetics, National Research Institute for Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Chen Zhang
- Department of Genetics, National Research Institute for Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ruqian Ding
- Department of Genetics, National Research Institute for Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Erer Gao
- Department of Genetics, National Research Institute for Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Haiyan Luo
- Department of Genetics, National Research Institute for Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wei Liu
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Cailing Lu
- Department of Genetics, National Research Institute for Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
2
|
Du G, Yan Y, Gao JF, Guo CY, Shen X, Lei XW. Therapeutic effect of folic acid combined with decitabine on diabetic mice. Int J Ophthalmol 2023; 16:1766-1772. [PMID: 38028519 PMCID: PMC10626348 DOI: 10.18240/ijo.2023.11.05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 08/15/2023] [Indexed: 12/01/2023] Open
Abstract
AIM To evaluate the therapeutic effect of folic acid combined with decitabine on diabetic mice. METHODS The diabetic model of db/db mice were randomly divided into model group, folic acid group, decitabine group, folic acid combined with decitabine group, and C57 mice as normal control group. The density of retinal blood vessels and retinal thickness were detected by fundus photography and optical coherence tomography, respectively. Pathological changes of retina were observed by hematoxylin-eosin (HE) staining. The homocysteine (Hcy) in serum was detected by enzyme linked immunosorbent assay (ELISA). TdT-mediated dUTP nick-end labeling (TUNEL) was used to detect apoptosis in retinal tissue. Evans blue dye was used to detect the permeability of retinal blood vessels. The platelet endothelial cell adhesion molecule-1 (CD31) and vascular endothelial growth factor receptor (VEGFR) protein were detected by Western blot. The 3-nitrotyrosine (3-NT) and 4-hydroxynonanine (4-HNE) were detected by immunohistochemistry. RESULTS The density of retinal blood vessels, retinal thickness, retinal vascular permeability and the proportion of apoptotic cells of retinal tissue in the model group increased significantly than control group (P<0.05). The Hcy in serum and the levels of CD31, VEGFR, 3-NT, and 4-HNE in retinal tissue increased significantly in the model group (P<0.01). Folic acid and decitabine both reversed these changes significantly, and the combination of the folic acid and decitabine worked best. CONCLUSION The combination of folic acid and decitabine has a more significant protective effect on the retina in diabetic mice.
Collapse
Affiliation(s)
- Gang Du
- The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Yong Yan
- Huining Second People's Hospital, Baiyin 730700, Gansu Province, China
| | - Jun-Feng Gao
- The Fourth People's Hospital of Tianshui, Tianshui 741000, Gansu Province, China
| | - Chun-Yan Guo
- Dingxi People's Hospital, Dingxi 743000, Gansu Province, China
| | - Xiao Shen
- The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Xun-Wen Lei
- The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
3
|
Zhao Y, E Z, Jiao A, Sun Z, Zhang H, Wang H, Fang N, Gao Q, Jin Q. Dendrobine enhances bovine oocyte maturation and subsequent embryonic development and quality. Theriogenology 2023; 203:53-60. [PMID: 36972665 DOI: 10.1016/j.theriogenology.2023.03.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 03/14/2023] [Accepted: 03/14/2023] [Indexed: 03/28/2023]
Abstract
Strategies for improving the quality of oocytes have important theoretical and practical significance for increasing the efficiency of livestock breeding. In this respect, the accumulation of reactive oxygen species (ROS) is a major factor affecting the development of oocytes and embryos. This study investigated the effects of Dendrobium nobile extract (DNE) on the in vitro maturation of bovine oocytes and embryonic development after IVF. DNE is an extract from Dendrobium rhizomes that contains alkaloids with anti-inflammatory, anti-cancer and anti-ageing functions. Various concentrations of DNE (0, 5, 10, 20 and 50 μmol/L) were added during oocyte maturation in vitro, and we found that 10 μmol/L of DNE remarkably increased the oocyte maturation rate, the subsequent blastocyst formation rate and embryo quality. Further, we found that DNE treatment decreased the frequency of spindle/chromosome defects and ROS and increased the oocyte glutathione and mitochondrial membrane potential in oocytes. Moreover, DNE upregulated the expression of oxidative stress-related genes (Sirt1, Sirt2, Sirt3 and Sod1) in oocytes and apoptosis-related genes (Caspase-3, Caspase-4, Bax, Bcl-xl and Survivin) in blastocysts. These results suggest that DNE supplementation can promote oocyte maturation and subsequent embryonic development by regulating redox reactions and inhibiting embryonic apoptosis.
Collapse
|
4
|
Wang Y, Jiang S, Chen X, Liu X, Li N, Nie Y, Lu G. Comparison of developmental toxicity of benzophenone-3 and its metabolite benzophenone-8 in zebrafish. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 258:106515. [PMID: 37011548 DOI: 10.1016/j.aquatox.2023.106515] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 06/19/2023]
Abstract
Benzophenone-3 (BP-3) as one of frequently used organic UV filters has been considered an emerging pollutant due to its toxicities. Benzophenone-8 (BP-8) is one of the main metabolites of BP-3 in organisms. Current reports show that BP-8 may be more toxic than BP-3. However, difference of their toxicities on embryonic development has rarely been reported. In this study, zebrafish embryos were chosen as the target organism to explore the developmental toxicities of BP-3 and BP-8. Non-targeted metabolomic analysis was performed to compare their modes of action. Results showed that BP-8 exposures led to higher bioaccumulation and lower hatching rate of zebrafish larvae than BP-3. Both BP-8 and BP-3 exposures caused behavioral abnormalities of zebrafish larvae, but no significant difference was found between them. At the metabolome level, 1 μg/L BP-3 and 1 μg/L BP-8 exposures altered neuroactive ligand-receptor interaction pathway and FoxO signaling pathway, respectively, which might be involved in the abnormal behaviors in zebrafish larvae. For higher exposure groups (30 and 300 μg/L), both BP-3 and BP-8 exposures changed metabolism of cofactors and vitamins of zebrafish larvae. Exposure of BP-3 altered the metabolism by pantothenate and CoA biosynthesis pathway, while BP-8 exposure changed riboflavin metabolism and folate biosynthesis. The above results indicated different modes of action of BP-3 and BP-8 in zebrafish embryonic development. This study sheds new light to biological hazards of BP-3 due to its metabolism in aquatic organisms.
Collapse
Affiliation(s)
- Yonghua Wang
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lake of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, PR China.
| | - Shengnan Jiang
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lake of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, PR China
| | - Xi Chen
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lake of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, PR China
| | - Xiaodan Liu
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lake of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, PR China
| | - Na Li
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lake of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, PR China
| | - Yang Nie
- Hangzhou Hydrology and Water Resources Monitoring Center, Hangzhou 310016, PR China.
| | - Guanghua Lu
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lake of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, PR China
| |
Collapse
|
5
|
Zhang Y, Mustieles V, Sun Q, Coull B, McElrath T, Rifas-Shiman SL, Martin L, Sun Y, Wang YX, Oken E, Cardenas A, Messerlian C. Association of Early Pregnancy Perfluoroalkyl and Polyfluoroalkyl Substance Exposure With Birth Outcomes. JAMA Netw Open 2023; 6:e2314934. [PMID: 37256622 PMCID: PMC10233420 DOI: 10.1001/jamanetworkopen.2023.14934] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 04/10/2023] [Indexed: 06/01/2023] Open
Abstract
Importance Prenatal perfluoroalkyl and polyfluoroalkyl substances (PFAS) have been linked to adverse birth outcomes. Previous research showed that higher folate concentrations are associated with lower blood PFAS concentrations in adolescents and adults. Further studies are needed to explore whether prenatal folate status mitigates PFAS-related adverse birth outcomes. Objective To examine whether prenatal folate status modifies the negative associations between pregnancy PFAS concentrations, birth weight, and gestational age previously observed in a US cohort. Design, Setting, and Participants In a prospective design, a prebirth cohort of mothers or pregnant women was recruited between April 1999 and November 2002, in Project Viva, a study conducted in eastern Massachusetts. Statistical analyses were performed from May 24 and October 25, 2022. Exposure Plasma concentrations of 6 PFAS compounds were measured in early pregnancy (median gestational week, 9.6). Folate status was assessed through a food frequency questionnaire and measured in plasma samples collected in early pregnancy. Main Outcomes and Measures Birth weight and gestational age, abstracted from delivery records; birth weight z score, standardized by gestational age and infant sex; low birth weight, defined as birth weight less than 2500 g; and preterm birth, defined as birth at less than 37 completed gestational weeks. Results The cohort included a total of 1400 mother-singleton pairs. The mean (SD) age of the mothers was 32.21 (4.89) years. Most of the mothers were White (73.2%) and had a college degree or higher (69.1%). Early pregnancy plasma perfluorooctanoic acid concentration was associated with lower birth weight and birth weight z score only among mothers whose dietary folate intake (birth weight: β, -89.13 g; 95% CI, -166.84 to -11.42 g; birth weight z score: -0.13; 95% CI, -0.26 to -0.003) or plasma folate concentration (birth weight: -87.03 g; 95% CI, -180.11 to 6.05 g; birth weight z score: -0.14; 95% CI, -0.30 to 0.02) were below the 25th percentile (dietary: 660 μg/d, plasma: 14 ng/mL). No associations were found among mothers in the higher folate level groups, although the tests for heterogeneity did not reject the null. Associations between plasma perfluorooctane sulfonic acid and perfluorononanoate (PFNA) concentrations and lower birth weight, and between PFNA and earlier gestational age were noted only among mothers whose prenatal dietary folate intake or plasma folate concentration was in the lowest quartile range. No associations were found among mothers in higher folate status quartile groups. Conclusions and Relevance In this large, US prebirth cohort, early pregnancy exposure to select PFAS compounds was associated with adverse birth outcomes only among mothers below the 25th percentile of prenatal dietary or plasma folate levels.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Vicente Mustieles
- University of Granada, Center for Biomedical Research, Instituto de Investigación Biosanitaria Ibs, Consortium for Biomedical Research in Epidemiology and Public Health Grenada, Spain
| | - Qi Sun
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Brent Coull
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Thomas McElrath
- Harvard Medical School, Boston, Massachusetts
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Sheryl L. Rifas-Shiman
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts
| | - Leah Martin
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Yang Sun
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Yi-Xin Wang
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Emily Oken
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts
| | - Andres Cardenas
- Department of Epidemiology and Population Health, Stanford University, Stanford, California
| | - Carmen Messerlian
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital Fertility Center, Boston
| |
Collapse
|
6
|
Simonian R, Pannia E, Hammoud R, Noche RR, Cui X, Kranenburg E, Kubant R, Ashcraft P, Wasek B, Bottiglieri T, Dowling JJ, Anderson GH. Methylenetetrahydrofolate reductase deficiency and high-dose FA supplementation disrupt embryonic development of energy balance and metabolic homeostasis in zebrafish. Hum Mol Genet 2023; 32:1575-1588. [PMID: 36637428 PMCID: PMC10117162 DOI: 10.1093/hmg/ddac308] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/03/2022] [Accepted: 12/22/2022] [Indexed: 01/14/2023] Open
Abstract
Folic acid (synthetic folate, FA) is consumed in excess in North America and may interact with common pathogenic variants in methylenetetrahydrofolate reductase (MTHFR); the most prevalent inborn error of folate metabolism with wide-ranging obesity-related comorbidities. While preclinical murine models have been valuable to inform on diet-gene interactions, a recent Folate Expert panel has encouraged validation of new animal models. In this study, we characterized a novel zebrafish model of mthfr deficiency and evaluated the effects of genetic loss of mthfr function and FA supplementation during embryonic development on energy homeostasis and metabolism. mthfr-deficient zebrafish were generated using CRISPR mutagenesis and supplemented with no FA (control, 0FA) or 100 μm FA (100FA) throughout embryonic development (0-5 days postfertilization). We show that the genetic loss of mthfr function in zebrafish recapitulates key biochemical hallmarks reported in MTHFR deficiency in humans and leads to greater lipid accumulation and aberrant cholesterol metabolism as reported in the Mthfr murine model. In mthfr-deficient zebrafish, energy homeostasis was also impaired as indicated by altered food intake, reduced metabolic rate and lower expression of central energy-regulatory genes. Microglia abundance, involved in healthy neuronal development, was also reduced. FA supplementation to control zebrafish mimicked many of the adverse effects of mthfr deficiency, some of which were also exacerbated in mthfr-deficient zebrafish. Together, these findings support the translatability of the mthfr-deficient zebrafish as a preclinical model in folate research.
Collapse
Affiliation(s)
- Rebecca Simonian
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Emanuela Pannia
- Department of Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Rola Hammoud
- Department of Laboratory Medicine and Pathobiology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto ON, M5G 1X5, Canada
| | - Ramil R Noche
- Department of Comparative Medicine, Yale Zebrafish Research Core, Yale School of Medicine, New Haven, CT 06511, USA
| | - Xiucheng Cui
- Department of Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Eva Kranenburg
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ruslan Kubant
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Paula Ashcraft
- Baylor Scott & White Research Institute, Institute of Metabolic Disease, Dallas, TX 75204, USA
| | - Brandi Wasek
- Baylor Scott & White Research Institute, Institute of Metabolic Disease, Dallas, TX 75204, USA
| | - Teodoro Bottiglieri
- Baylor Scott & White Research Institute, Institute of Metabolic Disease, Dallas, TX 75204, USA
| | - James J Dowling
- Department of Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - G Harvey Anderson
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
7
|
Zhang C, Dan Q, Lai S, Zhang Y, Gao E, Luo H, Yang L, Gao X, Lu C. Rab10 protects against DOX-induced cardiotoxicity by alleviating the oxidative stress and apoptosis of cardiomyocytes. Toxicol Lett 2023; 373:84-93. [PMID: 36309171 DOI: 10.1016/j.toxlet.2022.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 09/28/2022] [Accepted: 10/24/2022] [Indexed: 11/07/2022]
Abstract
Doxorubicin (DOX) is a widely used anticancer drug, but its clinical application is limited by cardiotoxicity. As a member of the Rab family, Rab10 has multiple subcellular localizations and carries out a wide variety of functions. Here, we explored the role of Rab10 on DOX-induced cardiotoxicity. Cardiac-specific Rab10 transgenic mice were constructed and treated with DOX or saline. We found that cardiac-specific overexpression of Rab10 alleviated cardiac dysfunction and attenuated cytoplasmic vacuolization and mitochondrial damage in DOX-treated mouse heart tissues. Immunofluorescence staining and Western blot analysis showed that Rab10 alleviated DOX-induced apoptosis and oxidative stress in cardiomyocytes in mouse heart tissues. We demonstrated that DOX mediated apoptosis, oxidative stress and depolarization of the mitochondrial membrane potential in H9c2 cells, while overexpression and knockdown of Rab10 attenuated and aggravated these effects, respectively. Furthermore, we found that Mst1, a serine-threonine kinase, was cleaved and translocated into the nucleus in H9c2 cells after DOX treatment, and knockdown of Mst1 alleviated DOX-induced cardiomyocyte apoptosis. Overexpression of Rab10 inhibited the cleavage of Mst1 mediated by DOX treatment in vivo and in vitro. Together, our findings demonstrated that cardiac-specific overexpression of Rab10 alleviated DOX-induced cardiac dysfunction and injury via inhibiting oxidative stress and apoptosis of cardiomyocytes, which may be partially ascribed to the inhibition of Mst1 activity.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Genetics, National Research Institute for Family Planning, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China
| | - Qinghua Dan
- Department of Genetics, National Research Institute for Family Planning, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China
| | - Song Lai
- Department of Cardiology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Yutong Zhang
- Department of Genetics, National Research Institute for Family Planning, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China
| | - Erer Gao
- Department of Genetics, National Research Institute for Family Planning, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China
| | - Haiyan Luo
- Department of Genetics, National Research Institute for Family Planning, Beijing, China
| | - Liping Yang
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, China
| | - Xiaobo Gao
- Department of Genetics, National Research Institute for Family Planning, Beijing, China.
| | - Cailing Lu
- Department of Genetics, National Research Institute for Family Planning, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China.
| |
Collapse
|
8
|
Je JG, Jiang Y, Heo JH, Li X, Jeon YJ, Ryu BM. Mitigative Effects of PFF-A Isolated from Ecklonia cava on Pigmentation in a Zebrafish Model and Melanogenesis in B16F10 Cells. Mar Drugs 2022; 20:123. [PMID: 35200653 PMCID: PMC8877154 DOI: 10.3390/md20020123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 01/29/2022] [Accepted: 02/03/2022] [Indexed: 02/01/2023] Open
Abstract
Melanin synthesis is a defense mechanism that prevents skin damage, but excessive accumulation of melanin occurs in the skin in various reactions such as pigmentation, lentigines, and freckles. Although anti-melanogenic effects have been demonstrated for various naturally occurring marine products that inhibit and control tyrosinase activity, most studies have not been extended to in vivo applications. Phlorofucofuroeckol-A (PFF-A, 12.5-100 µM) isolated from Ecklonia cava has previously been shown to have tyrosinase-mitigative effects in B16F10 cells, but it has not been evaluated in an in vivo model, and its underlying mechanism for anti-melanogenic effects has not been studied. In the present study, we evaluated the safety and efficacy of PFF-A for anti-melanogenic effects in an in vivo model. We selected low doses of PFF-A (1.5-15 nM) and investigated their mitigative effects on pigmentation stimulated by α-MSH in vivo and their related-mechanism in an in vitro model. The findings suggest that low-dose PFF-A derived from E. cava suppresses pigmentation in vivo and melanogenesis in vitro. Therefore, this study presents the possibility that PFF-A could be utilized as a new anti-melanogenic agent in the cosmeceutical industries.
Collapse
Affiliation(s)
- Jun-Geon Je
- Department of Marine Life Sciences, Jeju National University, Jeju 63243, Korea; (J.-G.J.); (J.-H.H.); (Y.-J.J.)
| | - Yunfei Jiang
- School of Food Engineering, Jilin Agriculture Science and Technology University, Jilin 132101, China;
| | - Jun-Ho Heo
- Department of Marine Life Sciences, Jeju National University, Jeju 63243, Korea; (J.-G.J.); (J.-H.H.); (Y.-J.J.)
| | - Xining Li
- School of Life Sciences, Northeast Normal University, Changchun 130024, China;
| | - You-Jin Jeon
- Department of Marine Life Sciences, Jeju National University, Jeju 63243, Korea; (J.-G.J.); (J.-H.H.); (Y.-J.J.)
- Marine Science Institute, Jeju National University, Jeju 63333, Korea
| | - Bo-Mi Ryu
- Department of Marine Life Sciences, Jeju National University, Jeju 63243, Korea; (J.-G.J.); (J.-H.H.); (Y.-J.J.)
| |
Collapse
|
9
|
Sijko M, Kozłowska L. Influence of Dietary Compounds on Arsenic Metabolism and Toxicity. Part II-Human Studies. TOXICS 2021; 9:259. [PMID: 34678956 PMCID: PMC8541625 DOI: 10.3390/toxics9100259] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 09/25/2021] [Indexed: 01/25/2023]
Abstract
Exposure to various forms of arsenic (As), the source of which may be environmental as well as occupational exposure, is associated with many adverse health effects. Therefore, methods to reduce the adverse effects of As on the human body are being sought. Research in this area focuses, among other topics, on the dietary compounds that are involved in the metabolism of this element. Therefore, the aim of this review was to analyze the influence of methionine, betaine, choline, folic acid, vitamin B2, B6, B12 and zinc on the efficiency of inorganic As (iAs) metabolism and the reduction in the severity of the whole spectrum of disorders related to As exposure. In this review, which included 62 original papers (human studies) we present the current knowledge in the area. In human studies, these compounds (methionine, choline, folic acid, vitamin B2, B6, B12 and zinc) may increase iAs metabolism and reduce toxicity, whereas their deficiency may impair iAs metabolism and increase As toxicity. Taking into account the results of studies conducted in populations exposed to As, it is reasonable to carry out prophylactic activities. In particular nutritional education seems to be important and should be focused on informing people that an adequate intake of those dietary compounds potentially has a modulating effect on iAs metabolism, thus, reducing its adverse effects on the body.
Collapse
Affiliation(s)
- Monika Sijko
- Department of Dietetics, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences (SGGW-WULS), 159c Nowoursynowska Street, 02-776 Warsaw, Poland
| | - Lucyna Kozłowska
- Department of Dietetics, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences (SGGW-WULS), 159c Nowoursynowska Street, 02-776 Warsaw, Poland
| |
Collapse
|
10
|
Sijko M, Kozłowska L. Influence of Dietary Compounds on Arsenic Metabolism and Toxicity. Part I-Animal Model Studies. TOXICS 2021; 9:toxics9100258. [PMID: 34678954 PMCID: PMC8536957 DOI: 10.3390/toxics9100258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/17/2021] [Accepted: 09/25/2021] [Indexed: 12/14/2022]
Abstract
Population and laboratory studies indicate that exposure to various forms of arsenic (As) is associated with many adverse health effects; therefore, methods are being sought out to reduce them. Numerous studies focus on the effects of nutrients on inorganic As (iAs) metabolism and toxicity, mainly in animal models. Therefore, the aim of this review was to analyze the influence of methionine, betaine, choline, folic acid, vitamin B2, B6, B12 and zinc on the efficiency of iAs metabolism and the reduction of the severity of the whole spectrum of disorders related to iAs exposure. In this review, which includes 58 (in vivo and in vitro studies) original papers, we present the current knowledge in the area. In vitro and in vivo animal studies showed that methionine, choline, folic acid, vitamin B2, B12 and zinc reduced the adverse effects of exposure to iAs in the gastrointestinal, urinary, lymphatic, circulatory, nervous, and reproductive systems. On the other hand, it was observed that these compounds (methionine, choline, folic acid, vitamin B2, B12 and zinc) may increase iAs metabolism and reduce toxicity, whereas their deficiency or excess may impair iAs metabolism and increase iAs toxicity. Promising results of in vivo and in vitro on animal model studies show the possibility of using these nutrients in populations particularly exposed to As.
Collapse
Affiliation(s)
- Monika Sijko
- Correspondence: (M.S.); (L.K.); Tel.: +48-22-59-370-23 (M.S.); +48-22-59-370-17 (L.K.)
| | - Lucyna Kozłowska
- Correspondence: (M.S.); (L.K.); Tel.: +48-22-59-370-23 (M.S.); +48-22-59-370-17 (L.K.)
| |
Collapse
|
11
|
Muhsen M, Youngs J, Riu A, Gustafsson JÅ, Kondamadugu VS, Garyfalidis E, Bondesson M. Folic acid supplementation rescues valproic acid-induced developmental neurotoxicity and behavioral alterations in zebrafish embryos. Epilepsia 2021; 62:1689-1700. [PMID: 33997963 DOI: 10.1111/epi.16915] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Fetal exposure to the anticonvulsant drug valproic acid (VPA), used to treat certain types of epilepsy, increases the risk for birth defects, including neural tube defects, as well as learning difficulties and behavioral problems. Here, we investigated neurotoxic effects of VPA exposure using zebrafish as a model organism. The capacity of folic acid (FA) supplementation to rescue the VPA-induced neuronal and behavioral perturbations was also examined. METHODS Zebrafish embryos of different transgenic lines with neuronal green fluorescent protein expression were exposed to increasing concentrations of VPA with or without FA supplementation. Fluorescence microscopy was used to visualize alterations in brain structures and neural progenitor cells, as well as motor neurons and neurite sprouting. A twitching behavioral assay was used to examine the functional consequences of VPA and FA treatment. RESULTS In zebrafish embryos, VPA exposure caused a decrease in the midbrain size, an increase in the midline gap of the hindbrain, and perturbed neurite sprouting of secondary motor neurons, in a concentration-dependent manner. VPA exposure also decreased the fluorescence intensity of neuronal progenitor cells in early developmental stages, indicating fewer cells. Furthermore, VPA exposure significantly altered embryonic twitching activity, causing hyperactivity in dark and hypoactivity in light. Supplementation of FA rescued the VPA-induced smaller midbrain size and hindbrain midline gap defects. FA treatment also increased the number of neuronal progenitor cells in VPA-treated embryos and salvaged neurite sprouting of the secondary motor neurons. FA rescued the VPA-induced alterations in twitching activity in light but not in dark. SIGNIFICANCE We conclude that VPA exposure induces specific neurotoxic perturbations in developing zebrafish embryos, and that FA reversed most of the identified defects. The results demonstrate that zebrafish is a promising model to study VPA-induced teratogenesis and to screen for countermeasures.
Collapse
Affiliation(s)
- Maram Muhsen
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana, USA
| | - Jaclyn Youngs
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana, USA
| | - Anne Riu
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas, USA
| | - Jan-Åke Gustafsson
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas, USA.,Department of Biosciences and Nutrition, Karolinska Institute, Solna, Sweden
| | - Vijay Sai Kondamadugu
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana, USA
| | - Elefterios Garyfalidis
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana, USA
| | - Maria Bondesson
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, Indiana, USA
| |
Collapse
|
12
|
Gao X, Zhang C, Zheng P, Dan Q, Luo H, Ma X, Lu C. Arsenic suppresses GDF1 expression via ROS-dependent downregulation of specificity protein 1. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 271:116302. [PMID: 33360347 DOI: 10.1016/j.envpol.2020.116302] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/06/2020] [Accepted: 12/10/2020] [Indexed: 06/12/2023]
Abstract
Inorganic arsenic, an environmental contaminant, has adverse health outcomes. Our previous studies showed that arsenic causes abnormal cardiac development in zebrafish embryos by downregulating Dvr1/GDF1 expression and that folic acid protects against these effects. However, the mechanism by which arsenic represses Dvr1/GDF1 expression remains unknown. Herein, we demonstrate that specificity protein 1 (Sp1) acts as a transcriptional activator of GDF1. Arsenic treatment downregulated Sp1 at both the mRNA and protein level and its downstream targets GDF1 and SIRT1. Chromatin immunoprecipitation analysis showed that the occupancy of Sp1 on the GDF1 or SIRT1 promoter was significantly reduced in response to arsenite. Further investigation showed that Sp1 overexpression inhibited the arsenic-mediated decrease in GDF1 and SIRT1, while Sp1 knockdown had the opposite effect. We found that expression of the oxidative adaptor p66shc was inversely related to that of SIRT1 and that the binding of SIRT1 to the p66shc promoter was sharply attenuated by arsenite treatment. SIRT1 overexpression attenuated p66shc expression but enhanced GDF1 protein expression, while SIRT1 depletion exerted the opposite effect. Both the antioxidants N-acetylcysteine and folic acid reversed the arsenic-mediated repression of Sp1, GDF1 and SIRT1. Moreover, wild-type p66shc overexpression enhanced the arsenic-mediated repression of Sp1, GDF1 and SIRT1, which was accompanied by an increase in intracellular reactive oxygen species (ROS) levels, while both overexpression of a dominant negative p66shcSer36Ala mutant and deficiency in p66shc reversed these effects. Taken together, our results revealed that arsenic suppresses GDF1 expression via the ROS-dependent downregulation of the Sp1/SIRT1 axis, which forms a negative feedback loop with p66shc to regulate oxidative stress. Our findings reveal a novel molecular mechanism underlying arsenic toxicity and provide new insight into the protective effect of folic acid in arsenic-mediated toxicity.
Collapse
Affiliation(s)
- Xiaobo Gao
- Department of Genetics, National Research Institute for Family Planning, Beijing, China
| | - Chen Zhang
- Department of Genetics, National Research Institute for Family Planning, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China
| | - Panpan Zheng
- Department of Genetics, National Research Institute for Family Planning, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China
| | - Qinghua Dan
- Department of Genetics, National Research Institute for Family Planning, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China
| | - Haiyan Luo
- Department of Genetics, National Research Institute for Family Planning, Beijing, China
| | - Xu Ma
- Department of Genetics, National Research Institute for Family Planning, Beijing, China
| | - Cailing Lu
- Department of Genetics, National Research Institute for Family Planning, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China.
| |
Collapse
|
13
|
Delaney P, Ramdas Nair A, Palmer C, Khan N, Sadler KC. Arsenic induced redox imbalance triggers the unfolded protein response in the liver of zebrafish. Toxicol Appl Pharmacol 2020; 409:115307. [PMID: 33147493 DOI: 10.1016/j.taap.2020.115307] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/01/2020] [Accepted: 10/26/2020] [Indexed: 12/16/2022]
Abstract
Inorganic arsenic (iAs) is one of the most endemic toxicants worldwide and oxidative stress is a key cellular pathway underlying iAs toxicity. Other cellular stress response pathways, such as the unfolded protein response (UPR), are also impacted by iAs exposure, however it is not known how these pathways intersect to cause disease. We optimized the use of zebrafish larvae to identify the relationship between these cellular stress response pathways and arsenic toxicity. We found that the window of iAs susceptibility during zebrafish development corresponds with the development of the liver, and that even a 24-h exposure can cause lethality if administered to mature larvae, but not to early embryos. Acute exposure of larvae to iAs generates reactive oxygen species (ROS), an antioxidant response, endoplasmic reticulum (ER) stress and UPR activation in the liver. An in vivo assay using transgenic larvae expressing a GFP-tagged secreted glycoprotein in hepatocytes (Tg(fabp10a:Gc-EGFP)) revealed acute iAs exposure selectively decreased expression of Gc-EGFP, indicating that iAs impairs secretory protein folding in the liver. The transcriptional output of UPR activation is preceded by ROS production and activation of genes involved in the oxidative stress response. These studies implicate redox imbalance as the mechanism of iAs-induced ER stress and suggest that crosstalk between these pathways underlie iAs-induced hepatic toxicity.
Collapse
Affiliation(s)
- Patrice Delaney
- Program in Biology, New York University Abu Dhabi, Saadiyat Island, United Arab Emirates
| | - Anjana Ramdas Nair
- Program in Biology, New York University Abu Dhabi, Saadiyat Island, United Arab Emirates
| | - Catherine Palmer
- Program in Biology, New York University Abu Dhabi, Saadiyat Island, United Arab Emirates
| | - Nouf Khan
- Program in Biology, New York University Abu Dhabi, Saadiyat Island, United Arab Emirates
| | - Kirsten C Sadler
- Program in Biology, New York University Abu Dhabi, Saadiyat Island, United Arab Emirates.
| |
Collapse
|
14
|
Sun HJ, Zhang JY, Wang Q, Zhu E, Chen W, Lin H, Chen J, Hong H. Environmentally relevant concentrations of arsenite induces developmental toxicity and oxidative responses in the early life stage of zebrafish. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 254:113022. [PMID: 31408795 DOI: 10.1016/j.envpol.2019.113022] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 07/15/2019] [Accepted: 08/04/2019] [Indexed: 06/10/2023]
Abstract
Arsenic (As) present in water is a nonignorable environmental issue, even at low concentrations (≤150 μg L-1). To evaluate the toxic effect of low concentrations of As, zebrafish at early life stage were exposed to 0, 25, 50, 75, or 150 μg L-1 AsIII for 120 h. Our results indicated that low concentration of AsIII decreased zebrafish larvae's survival rate to 85%, 89% and 86% at 50, 75 and 150 μg L-1. Furthermore, low concentrations of AsIII exposure caused oxidative stress (elevated superoxide dismutase (SOD) activity and influenced the mRNA transcriptional levels of Cu/ZnSOD and MnSOD) and damage (increased malondialdehyde levels). Meanwhile, zebrafish larvae regulated the mRNA transcription of metallothionein and heat shock protein 70 to alleviate toxicity caused by AsIII. These results revealed lower concentrations (≤150 μg L-1) of AsIII had a detriment effect on the survival of fish at early life stage, moreover, oxidative stress caused by AsIII posed potential risk for the zebrafish. This study provides novel insight into low concentration AsIII-induced toxicity in zebrafish.
Collapse
Affiliation(s)
- Hong-Jie Sun
- College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang 321004, China.
| | - Jing-Ying Zhang
- College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang 321004, China
| | - Qiang Wang
- College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang 321004, China
| | - Engao Zhu
- College of Chemistry and Life Science, Zhejiang Normal University, Jinhua, Zhejiang 321004, China
| | - Wenrong Chen
- College of Chemistry and Life Science, Zhejiang Normal University, Jinhua, Zhejiang 321004, China
| | - Hongjun Lin
- College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang 321004, China
| | - Jianrong Chen
- College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang 321004, China
| | - Huachang Hong
- College of Geography and Environmental Science, Zhejiang Normal University, Jinhua, Zhejiang 321004, China
| |
Collapse
|
15
|
Wong CP, Dashner-Titus EJ, Alvarez SC, Chase TT, Hudson LG, Ho E. Zinc Deficiency and Arsenic Exposure Can Act Both Independently or Cooperatively to Affect Zinc Status, Oxidative Stress, and Inflammatory Response. Biol Trace Elem Res 2019; 191:370-381. [PMID: 30635848 PMCID: PMC6625954 DOI: 10.1007/s12011-019-1631-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/01/2019] [Indexed: 12/16/2022]
Abstract
The negative health impact of zinc deficiency overlaps significantly with arsenic exposure, and is associated with increased risk for chronic diseases. Arsenic contamination in the groundwater often co-exists in regions of the world that are prone to zinc deficiency. Notably, low zinc status shares many hallmarks of arsenic exposure, including increased oxidative stress and inflammation. Despite their common targets and frequent co-distribution in the population, little is known regarding the interaction between zinc deficiency and arsenic exposure. In this study, we tested the effect of arsenic exposure at environmentally relevant doses in combination with a physiologically relevant level of zinc deficiency (marginal zinc deficiency) on zinc status, oxidative damage, and inflammation. In cell culture, zinc-deficient THP-1 monocytes co-exposed with arsenic resulted in further reduction in intracellular zinc, as well as further increase in oxidative stress and inflammatory markers. In an animal study, zinc-deficient mice had further decrease in zinc status when co-exposed to arsenic. Zinc deficiency, but not arsenic exposure, resulted in an increase in baseline transcript abundance of inflammatory markers in the liver. Upon lipopolysaccharide challenge to elicit an acute inflammatory response, arsenic exposure, but not zinc deficiency, resulted in an increase in proinflammatory response. In summary, zinc deficiency and arsenic exposure can function independently or cooperatively to affect zinc status, oxidant stress, and proinflammatory response. The results highlight the need to consider both nutritional status and arsenic exposures together when considering their impact on health outcomes in susceptible populations.
Collapse
Affiliation(s)
- Carmen P Wong
- School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Erica J Dashner-Titus
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Sandra C Alvarez
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Tyler T Chase
- School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - Laurie G Hudson
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Emily Ho
- School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR, 97331, USA.
- Linus Pauling Institute, Oregon State University, Corvallis, OR, 97331, USA.
- Moore Family Center for Whole Grain Foods, Nutrition and Preventive Health, Oregon State University, Corvallis, OR, 97331, USA.
| |
Collapse
|
16
|
Association of functional variant in GDF1 promoter with risk of congenital heart disease and its regulation by Nkx2.5. Clin Sci (Lond) 2019; 133:1281-1295. [PMID: 31171573 DOI: 10.1042/cs20181024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 05/13/2019] [Accepted: 06/06/2019] [Indexed: 11/17/2022]
Abstract
Abstract
GDF1 plays an important role in left–right patterning and genetic mutations in the coding region of GDF1 are associated with congenital heart disease (CHD). However, the genetic variation in the promoter of GDF1 with sporadic CHD and its expression regulation is little known. The association of the genetic variation in GDF1 promoter with CHD was examined in two case–control studies, including 1084 cases and 1198 controls in the first study and 582 cases and 615 controls in the second study. We identified one single nucleotide polymorphism (SNP) rs181317402 and two novel genetic mutations located in the promoter region of GDF1. Analysis of combined samples revealed a significant association in genotype and allele frequencies of rs181317402 T/G polymorphism between CHD cases in overall or ventricular septal defects or Tetralogy of Fallot and the control group. rs181317402 allele G polymorphism was significantly associated with a decreased risk of CHD. Furthermore, luciferase assay, chromatin immunoprecipitation and DNA pulldown assay indicated that Nkx2.5 transactivated the expression of GDF1 by binding to the promoter of GDF1. Luciferase activity assay showed that rs181317402 allele G significantly increased the basal and Nkx2.5-mediated activity of GDF1 promoter, while the two genetic mutations had the opposite effect. rs181317402 TG genotype was associated with significantly increased mRNA level of GDF1 compared with TT genotype in 18 CHD individuals. Our results demonstrate for the first time that Nkx2.5 acts upstream of GDF1 and the genetic variants in GDF1 promoter may confer genetic susceptibility to sporadic CHD potentially by altering its expression.
Collapse
|
17
|
Bambino K, Zhang C, Austin C, Amarasiriwardena C, Arora M, Chu J, Sadler KC. Inorganic arsenic causes fatty liver and interacts with ethanol to cause alcoholic liver disease in zebrafish. Dis Model Mech 2018; 11:dmm.031575. [PMID: 29361514 PMCID: PMC5894941 DOI: 10.1242/dmm.031575] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 12/07/2017] [Indexed: 12/19/2022] Open
Abstract
The rapid increase in fatty liver disease (FLD) incidence is attributed largely to genetic and lifestyle factors; however, environmental toxicants are a frequently overlooked factor that can modify the effects of more common causes of FLD. Chronic exposure to inorganic arsenic (iAs) is associated with liver disease in humans and animal models, but neither the mechanism of action nor the combinatorial interaction with other disease-causing factors has been fully investigated. Here, we examined the contribution of iAs to FLD using zebrafish and tested the interaction with ethanol to cause alcoholic liver disease (ALD). We report that zebrafish exposed to iAs throughout development developed specific phenotypes beginning at 4 days post-fertilization (dpf), including the development of FLD in over 50% of larvae by 5 dpf. Comparative transcriptomic analysis of livers from larvae exposed to either iAs or ethanol revealed the oxidative stress response and the unfolded protein response (UPR) caused by endoplasmic reticulum (ER) stress as common pathways in both these models of FLD, suggesting that they target similar cellular processes. This was confirmed by our finding that arsenic is synthetically lethal with both ethanol and a well-characterized ER-stress-inducing agent (tunicamycin), suggesting that these exposures work together through UPR activation to cause iAs toxicity. Most significantly, combined exposure to sub-toxic concentrations of iAs and ethanol potentiated the expression of UPR-associated genes, cooperated to induce FLD, reduced the expression of as3mt, which encodes an arsenic-metabolizing enzyme, and significantly increased the concentration of iAs in the liver. This demonstrates that iAs exposure is sufficient to cause FLD and that low doses of iAs can potentiate the effects of ethanol to cause liver disease. This article has an associated First Person interview with the first author of the paper. Summary: Using zebrafish, the authors show that exposure to a common environmental contaminant, inorganic arsenic, increases the risk of alcoholic liver disease.
Collapse
Affiliation(s)
- Kathryn Bambino
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Chi Zhang
- Program in Biology, New York University Abu Dhabi, Saadiyat Island Campus, PO Box 129188 Abu Dhabi, United Arab Emirates
| | - Christine Austin
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Chitra Amarasiriwardena
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Manish Arora
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Jaime Chu
- Department of Pediatrics, Division of Pediatric Hepatology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Kirsten C Sadler
- Program in Biology, New York University Abu Dhabi, Saadiyat Island Campus, PO Box 129188 Abu Dhabi, United Arab Emirates
| |
Collapse
|
18
|
Kurzius-Spencer M, da Silva V, Thomson CA, Hartz V, Hsu CH, Burgess JL, O'Rourke MK, Harris RB. Nutrients in one-carbon metabolism and urinary arsenic methylation in the National Health and Nutrition Examination Survey (NHANES) 2003-2004. THE SCIENCE OF THE TOTAL ENVIRONMENT 2017; 607-608:381-390. [PMID: 28697391 DOI: 10.1016/j.scitotenv.2017.07.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 07/01/2017] [Accepted: 07/03/2017] [Indexed: 05/28/2023]
Abstract
Exposure to inorganic arsenic (inAs), a potent toxicant, occurs primarily through ingestion of food and water. The efficiency with which it is methylated to mono and dimethyl arsenicals (MMA and DMA) affects toxicity. Folate, vitamins B12 and B6 are required for 1C metabolism, and studies have found that higher levels of these nutrients increase methylation capacity and are associated with protection against adverse health effects from inAs, especially in undernourished populations. Our aim was to determine whether 1C-related nutrients are associated with greater inAs methylation capacity in a general population sample with overall adequate nutrition and low levels of As exposure. Univariate and multivariable regression models were used to evaluate the relationship of dietary and blood nutrients to urinary As methylation in the National Health and Nutrition Examination Survey (NHANES) 2003-2004. Outcome variables were the percent of the sum of inAs and methylated As species (inAs+MMA+DMA) excreted as inAs, MMA, and DMA, and the ratio of MMA:DMA. In univariate models, dietary folate, vitamin B6 and protein intake were associated with lower urinary inAs% and greater DMA% in adults (≥18years), with similar trends in children (6-18). In adjusted models, vitamin B6 intake (p=0.011) and RBC folate (p=0.036) were associated with lower inAs%, while dietary vitamin B12 was associated with higher inAs% (p=0.002) and lower DMA% (p=0.030). Total plasma homocysteine was associated with higher MMA% (p=0.004) and lower DMA% (p=0.003), but not with inAs%; other blood nutrients showed no association with urinary As. Although effect size is small, these findings suggest that 1C nutrients can influence inAs methylation and potentially play an indirect role in reducing toxicity in a general population sample.
Collapse
Affiliation(s)
- Margaret Kurzius-Spencer
- Department of Pediatrics, College of Medicine, University of Arizona, Tucson, AZ, USA; Mel & Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA.
| | - Vanessa da Silva
- Department of Nutritional Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ, USA
| | - Cynthia A Thomson
- Mel & Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA; Department of Nutritional Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ, USA; The University of Arizona Cancer Center, Tucson, AZ, USA
| | - Vern Hartz
- The University of Arizona Cancer Center, Tucson, AZ, USA
| | - Chiu-Hsieh Hsu
- Mel & Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA; The University of Arizona Cancer Center, Tucson, AZ, USA
| | - Jefferey L Burgess
- Mel & Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
| | - Mary Kay O'Rourke
- Mel & Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
| | - Robin B Harris
- Mel & Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA; The University of Arizona Cancer Center, Tucson, AZ, USA
| |
Collapse
|
19
|
Rahman ML, Valeri L, Kile ML, Mazumdar M, Mostofa G, Qamruzzaman Q, Rahman M, Baccarelli A, Liang L, Hauser R, Christiani DC. Investigating causal relation between prenatal arsenic exposure and birthweight: Are smaller infants more susceptible? ENVIRONMENT INTERNATIONAL 2017; 108:32-40. [PMID: 28787626 PMCID: PMC5623127 DOI: 10.1016/j.envint.2017.07.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 06/03/2017] [Accepted: 07/30/2017] [Indexed: 05/21/2023]
Abstract
BACKGROUND Shortening of gestation and intrauterine growth restriction (IUGR) are the two main determinants of birthweight. Low birthweight has been linked with prenatal arsenic exposure, but the causal relation between arsenic and birthweight is not well understood. OBJECTIVES We applied a quantile causal mediation analysis approach to determine the association between prenatal arsenic exposure and birthweight in relation to shortening of gestation and IUGR, and whether the susceptibility of arsenic exposure varies by infant birth sizes. METHODS In a longitudinal birth cohort in Bangladesh, we measured arsenic in drinking water (n=1182) collected at enrollment and maternal toenails (n=1104) collected ≤1-month postpartum using inductively coupled plasma mass spectrometry. Gestational age was determined using ultrasound at ≤16weeks' gestation. Demographic information was collected using a structured questionnaire. RESULTS Of 1184 singleton livebirths, 16.4% (n=194) were low birthweight (<2500g), 21.9% (n=259) preterm (<37weeks' gestation), and 9.2% (n=109) both low birthweight and preterm. The median concentrations of arsenic in drinking water and maternal toenails were 2.2μg/L (range: below the level of detection [LOD]-1400) and 1.2μg/g (range: <LOD-46.6), respectively. Prenatal arsenic exposure was negatively associated with birthweight, where the magnitude of the association varied across birthweight percentiles. The effect of arsenic on birthweight mediated via shortening of gestation affected all infants irrespective of birth sizes (β range: 10th percentile=-19.7g [95% CI: -26.7, -13.3] to 90th percentile=-10.9g [95% CI: -18.5, -5.9] per natural log water arsenic increase), whereas the effect via pathways independent of gestational age affected only the smaller infants (β range: 10th percentile=-28.0g [95% CI: -43.8, -9.9] to 20th percentile=-14.9g [95% CI: -30.3, -1.7] per natural log water arsenic increase). Similar pattern was observed for maternal toenail arsenic. CONCLUSIONS The susceptibility of prenatal arsenic exposure varied by infant birth sizes, placing smaller infants at greater risk of lower birthweight by shortening of gestation and possibly growth restriction. It is important to mitigate prenatal arsenic exposure to improve perinatal outcomes in Bangladesh.
Collapse
Affiliation(s)
- Mohammad L Rahman
- Harvard T.H. Chan School of Public Health, Department of Environmental Health, Boston, MA, USA
| | - Linda Valeri
- McLean Hospital, Belmont, Massachusetts, USA, Harvard Medical School, Boston, MA, USA
| | - Molly L Kile
- Oregon State University, College of Public Health and Human Sciences, Corvallis, OR, USA
| | - Maitreyi Mazumdar
- Harvard T.H. Chan School of Public Health, Department of Environmental Health, Boston, MA, USA
| | | | | | | | - Andrea Baccarelli
- Columbia University, Mailman School of Public Health, Department of Environmental Health, New York, NY, USA
| | - Liming Liang
- Harvard T.H. Chan School of Public Health, Department of Biostatistics, Boston, MA, USA
| | - Russ Hauser
- Harvard T.H. Chan School of Public Health, Department of Environmental Health, Boston, MA, USA; Harvard T.H. Chan School of Public Health, Department of Epidemiology, Boston, MA, USA
| | - David C Christiani
- Harvard T.H. Chan School of Public Health, Department of Environmental Health, Boston, MA, USA; Harvard T.H. Chan School of Public Health, Department of Epidemiology, Boston, MA, USA.
| |
Collapse
|
20
|
Yue C, Ji C, Zhang H, Zhang LW, Tong J, Jiang Y, Chen T. Protective effects of folic acid on PM2.5-induced cardiac developmental toxicity in zebrafish embryos by targeting AhR and Wnt/β-catenin signal pathways. ENVIRONMENTAL TOXICOLOGY 2017; 32:2316-2322. [PMID: 28722335 DOI: 10.1002/tox.22448] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 06/21/2017] [Accepted: 07/02/2017] [Indexed: 05/06/2023]
Abstract
Our previous observations indicated that extractable organic matter (EOM) from PM2.5 induced malformations in the heart of zebrafish embryos by activating AhR and inhibiting canonical Wnt/β-catenin signal pathway. As a nutritional factor, folic acid (FA) is reported to prevent cardiac defects during embryo development. Hence, we hypothesize that FA may prevent PM2.5-induced heart defects by interfering with AhR and Wnt/β-catenin signaling pathways. Our results showed that FA supplementation alleviated the EOM-induced heart defects in zebrafish embryos, and both AhR inhibitor CH223191 and Wnt activator CHIR99021 enhanced the protective efficiency of FA. Furthermore, FA supplementation attenuated the EOM-induced upregulation of AhR and its target genes including Cyp1a1, Cyp1b1, Ahrra, and Ahrrb. EROD assay confirmed that the EOM agonized Cyp1a1 activity was diminished by FA. The EOM-induced downregulation of β-catenin and its target genes including Nkx2.5, Axin2, Sox9b, and Cox2b were recovered or even overexpressed in embryos exposed to EOM plus FA. In conclusion, our study suggested that FA supplementation protected against PM2.5 cardiac development toxicity by targeting AhR and Wnt/β-catenin signal pathways.
Collapse
Affiliation(s)
- Cong Yue
- Department of Toxicology, School of Public Health, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Cheng Ji
- Department of Genetics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Hang Zhang
- Department of Toxicology, School of Public Health, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Leshuai W Zhang
- School for Radiological and interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Jian Tong
- Department of Toxicology, School of Public Health, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Yan Jiang
- Department of Physiology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Tao Chen
- Department of Toxicology, School of Public Health, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| |
Collapse
|
21
|
Paul S, Bhattacharjee P, Giri AK, Bhattacharjee P. Arsenic toxicity and epimutagenecity: the new LINEage. Biometals 2017; 30:505-515. [PMID: 28516305 DOI: 10.1007/s10534-017-0021-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 05/09/2017] [Indexed: 12/15/2022]
Abstract
Global methylation pattern regulates the normal functioning of a cell. Research have shown arsenic alter these methylation landscapes within the genome leading to aberrant gene expression and inducts various pathophysiological outcomes. Long interspersed nuclear elements (LINE-1) normally remains inert due to heavy methylation of it's promoters, time and various environmental insults, they lose these methylation signatures and begin retro-transposition that has been associated with genomic instability and cancerous outcomes. Of the various high throughput technologies available to detect global methylation profile, development of LINE-1 methylation index shall provide a cost effect-screening tool to detect epimutagenic events in the wake of toxic exposure in a large number of individuals. In the present review, we tried to discuss the state of research and whether LINE-1 methylation can be considered as a potent epigenetic signature for arsenic toxicity.
Collapse
Affiliation(s)
- Somnath Paul
- Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Kolkata, 700032, India. .,Department of Epigenetics & Molecular Carcinogenesis, The Virginia Harris Cockrell Cancer Center, The University of Texas, M.D. Anderson Cancer Center, Science Park, 1808 Park Road 1C, Smithville, TX, 78957, USA.
| | - Pritha Bhattacharjee
- Department of Environmental Science, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, India
| | - Ashok K Giri
- Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Kolkata, 700032, India.
| | - Pritha Bhattacharjee
- Department of Environmental Science, University of Calcutta, 35 Ballygunge Circular Road, Kolkata, 700019, India.
| |
Collapse
|
22
|
Abstract
As manufacturing processes and development of new synthetic compounds increase to keep pace with the expanding global demand, environmental health, and the effects of toxicant exposure are emerging as critical public health concerns. Additionally, chemicals that naturally occur in the environment, such as metals, have profound effects on human and animal health. Many of these compounds are in the news: lead, arsenic, and endocrine disruptors such as bisphenol A have all been widely publicized as causing disease or damage to humans and wildlife in recent years. Despite the widespread appreciation that environmental toxins can be harmful, there is limited understanding of how many toxins cause disease. Zebrafish are at the forefront of toxicology research; this system has been widely used as a tool to detect toxins in water samples and to investigate the mechanisms of action of environmental toxins and their related diseases. The benefits of zebrafish for studying vertebrate development are equally useful for studying teratogens. Here, we review how zebrafish are being used both to detect the presence of some toxins as well as to identify how environmental exposures affect human health and disease. We focus on areas where zebrafish have been most effectively used in ecotoxicology and in environmental health, including investigation of exposures to endocrine disruptors, industrial waste byproducts, and arsenic.
Collapse
Affiliation(s)
- Kathryn Bambino
- Icahn School of Medicine at Mount Sinai, New York, United States
| | - Jaime Chu
- Icahn School of Medicine at Mount Sinai, New York, United States.
| |
Collapse
|
23
|
Li C, Li P, Tan YM, Lam SH, Chan ECY, Gong Z. Metabolomic Characterizations of Liver Injury Caused by Acute Arsenic Toxicity in Zebrafish. PLoS One 2016; 11:e0151225. [PMID: 26967897 PMCID: PMC4788152 DOI: 10.1371/journal.pone.0151225] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 02/23/2016] [Indexed: 11/28/2022] Open
Abstract
Arsenic is one of the most common metalloid contaminants in groundwater and it has both acute and chronic toxicity affecting multiple organs. Details of the mechanism of arsenic toxicity are still lacking and profile studies at metabolic level are very limited. Using gas chromatography coupled with mass spectroscopy (GC/MS), we first generated metabolomic profiles from the livers of arsenic-treated zebrafish and identified 34 significantly altered metabolite peaks as potential markers, including four prominent ones: cholic acid, glycylglycine, glycine and hypotaurine. Combined results from GC/MS, histological examination and pathway analyses suggested a series of alterations, including apoptosis, glycogenolysis, changes in amino acid metabolism and fatty acid composition, accumulation of bile acids and fats, and disturbance in glycolysis related energy metabolism. The alterations in glycolysis partially resemble Warburg effect commonly observed in many cancer cells. However, cellular damages were not reflected in two conventional liver function tests performed, Bilirubin assay and alanine aminotransferase (ALT) assay, probably because the short arsenate exposure was insufficient to induce detectable damage. This study demonstrated that metabolic changes could reflect mild liver impairments induced by arsenic exposure, which underscored their potential in reporting early liver injury.
Collapse
Affiliation(s)
- Caixia Li
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Ping Li
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Yee Min Tan
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Siew Hong Lam
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- NUS Environmental Research Institute, National University of Singapore, Singapore, Singapore
| | - Eric C. Y. Chan
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Zhiyuan Gong
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- NUS Environmental Research Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|