Review Article

MicroRNA (miRNA) Differential Expression and Exposure to Crude-Oil- Related Compounds

Author(s): Gabriela Coronel Vargas*

Volume 10, Issue 2, 2021

Published on: 04 June, 2021

Page: [97 - 108] Pages: 12

DOI: 10.2174/2211536610666210604122131

open access plus

Abstract

This review summarizes studies on miRNA differential regulation related to exposure to crude oil and 20 different crude oil chemicals, such as hydrocarbons, sulphur, nitrogen, and metalcontaining compounds. It may be interesting to explore the possibility of using early post-transcriptional regulators as a potential novel exposure biomarker.

Crude oil has been defined as a highly complex mixture of solids, liquids, and gases. Given the toxicological properties of the petroleum components, its extraction and elaboration processes represent high-risk activities for the environment and human health, especially when accidental spills occur. The effects on human health of short-term exposure to petroleum are well known, but chronic exposure effects may variate depending on the exposure type (i.e., work, clean-up activities, or nearby residence).

As only two studies are focused on miRNA differential expression after crude-oil exposure, this review will also analyse the bibliography concerning different crude-oil or Petroleum-Related Compounds (PRC) exposure in Animalia L. kingdom and how it is related to differential miRNA transcript levels. Papers include in vitro, animal, and human studies across the world.

A list of 10 miRNAs (miR-142-5p, miR-126-3p, miR-24-3p, miR-451a, miR-16-5p, miR-28-5p, let-7b-5p, miR-320b, miR-27a-3p and miR-346) was created based on bibliography analysis and hypothesised as a possible “footprint” for crude-oil exposure. miRNA differential regulation can be considered a Big-Data related challenge, so different statistical programs and bioinformatics tools were used to have a better understanding of the biological significate of the most interesting data.

Keywords: MicroRNA, crude-oil, petroleum, environmental exposure, bioinformatics, petroleum-related compounds.

Graphical Abstract
[1]
Lichtveld M, Sherchan S, Gam KB, et al. The deepwater horizon oil spill through the lens of human health and the ecosystem. Curr Environ Health Rep 2016; 3(4): 370-8.
[http://dx.doi.org/10.1007/s40572-016-0119-7] [PMID: 27722880]
[2]
Eklund RL, Knapp LC, Sandifer PA, Colwell RC. Oil spills and human health: Contributions of the Gulf of Mexico research initiative. Geohealth 2019; 3(12): 391-406.
[http://dx.doi.org/10.1029/2019GH000217] [PMID: 32159026]
[3]
WHO. Occupational exposures in petroleum refining; crude oil and major petroleum fuels. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. IARC Monogr Eval Carcinog Risks Hum 1989; 45: 1-322. Available from: https://publications.iarc.fr/_publications/media/download/1627/13a3d47168725cce5807f8e876bc3677443e8b21.pdf [Accessed 22 October 2020]
[4]
Colvin KA, Lewis C, Galloway TS. Current issues confounding the rapid toxicological assessment of oil spills. Chemosphere 2020; 245: 125585.
[http://dx.doi.org/10.1016/j.chemosphere.2019.125585] [PMID: 31855760]
[5]
Ramirez MI, Arevalo AP, Sotomayor S, Bailon-Moscoso N. Contamination by oil crude extraction - Refinement and their effects on human health. Environ Pollut 2017; 231(1): 415-25.
[http://dx.doi.org/10.1016/j.envpol.2017.08.017] [PMID: 28826075]
[6]
O’Callaghan-Gordo C, Orta-Martínez M, Kogevinas M. Health effects of non-occupational exposure to oil extraction. Environ Health 2016; 15(1): 56.
[http://dx.doi.org/10.1186/s12940-016-0140-1] [PMID: 27117290]
[7]
Krishnamurthy J, Engel LS, Wang L, et al. Neurological symptoms associated with oil spill response exposures: Results from the deepwater horizon oil spill coast guard cohort study. Environ Int 2019; 131: 104963.
[http://dx.doi.org/10.1016/j.envint.2019.104963] [PMID: 31382236]
[8]
Olayinka OO, Adewusi AA, Olarenwaju OO, Aladesida AA. Concentration of polycyclic aromatic hydrocarbons and estimated human health risk of water samples around Atlas Cove, Lagos, Nigeria. J Health Pollut 2018; 8(20): 181210.
[http://dx.doi.org/10.5696/2156-9614-8.20.181210] [PMID: 30560009]
[9]
Maurice L, López F, Becerra S, et al. Drinking water quality in areas impacted by oil activities in Ecuador: Associated health risks and social perception of human exposure. Sci Total Environ 2019; 690: 1203-17.
[http://dx.doi.org/10.1016/j.scitotenv.2019.07.089] [PMID: 31470483]
[10]
Bebeteidoh OL, Kometa S, Pazouki K, Norman R. Sustained impact of the activities of local crude oil refiners on their host communities in Nigeria. Heliyon 2020; 6(6): e04000.
[http://dx.doi.org/10.1016/j.heliyon.2020.e04000] [PMID: 32529060]
[11]
Backes C, Meese E, Keller A. Specific miRNA Disease biomarkers in blood, serum and plasma: Challenges and prospects. Mol Diagn Ther 2016; 20(6): 509-18.
[http://dx.doi.org/10.1007/s40291-016-0221-4] [PMID: 27378479]
[12]
Xu T, Su N, Liu L, Zhang J, Wang H, Zhang W. miRBaseConverter: An R/bioconductor package for converting and retrieving miRNA name, accession, sequence and family information in different versions of miRBase. BMC Bioinformatics 2018; 19(19): 179-88.2018;
[http://dx.doi.org/10.1101/407148]
[13]
Pajak M, Simpson TI. MiRNAtap: MicroRNA targets - Aggregated predictions. Bioconductor 2020. Available from: https://bioconductor.org/packages/release/bioc/html/miRNAtap.html
[http://dx.doi.org/10.18129/B9.bioc.miRNAtap]
[14]
Alexa A, Rahnenfuhrer J. topGO: Enrichment analysis for gene ontology. R package 2020.
[http://dx.doi.org/10.18129/B9.bioc.topGO]
[15]
Supek F, Bošnjak M, Škunca N, Šmuc T. REVIGO Summarizes and visualizes long lists of gene ontology terms. PLoS One 2011; 6(7): 21800.
[http://dx.doi.org/10.1371/journal.pone.0021800]
[16]
Xu EG, Khursigara AJ, Li S, et al. mRNA-miRNA-seq reveals Neuro-cardio mechanisms of crude oil toxicity in red drum (Sciaenops ocellatus). Environ Sci Technol 2019; 53(6): 3296-305.
[http://dx.doi.org/10.1021/acs.est.9b00150] [PMID: 30816040]
[17]
Xu EG, Magnuson JT, Diamante G, et al. Changes in microRNA-mRNA signatures agree with morphological, physiological, and behavioral changes in larval mahi-mahi treated with deepwater horizon oil. Environ Sci Technol 2018; 52(22): 13501-10.
[http://dx.doi.org/10.1021/acs.est.8b04169] [PMID: 30376307]
[18]
Filippov SV, Yarushkin AA, Kalinina TS, Ovchinnikov VY, Knyazev RA, Gulyaeva LF. Effect of benzo(a)pyrene on the expression of miR-483-3p in hepatocyte primary culture and rat liver. Biochemistry (Mosc) 2019; 84(10): 1197-203.
[http://dx.doi.org/10.1134/S0006297919100080] [PMID: 31694515]
[19]
Brevik A, Lindeman B, Brunborg G, Duale N. Paternal benzo[a]pyrene exposure modulates microRNA expression patterns in the developing mouse embryo. Int J Cell Biol 2012; 2012: 407431.
[http://dx.doi.org/10.1155/2012/407431] [PMID: 22548065]
[20]
Zuo J, Brewer DS, Arlt VM, Cooper CS, Phillips DH. Benzo pyrene-induced DNA adducts and gene expression profiles in target and non-target organs for carcinogenesis in mice. BMC Genomics 2014; 15(1): 880.
[http://dx.doi.org/10.1186/1471-2164-15-880] [PMID: 25297811]
[21]
Malik AI, Williams A, Lemieux CL, White PA, Yauk CL. Hepatic mRNA, microRNA, and miR-34a-target responses in mice after 28 days exposure to doses of benzo(a)pyrene that elicit DNA damage and mutation. Environ Mol Mutagen 2012; 53(1): 10-21.
[http://dx.doi.org/10.1002/em.20668] [PMID: 21964900]
[22]
Malik DE, David RM, Gooderham NJ. Mechanistic evidence that benzo[a]pyrene promotes an inflammatory microenvironment that drives the metastatic potential of human mammary cells. Arch Toxicol 2018; 92(10): 3223-39.
[http://dx.doi.org/10.1007/s00204-018-2291-z] [PMID: 30155724]
[23]
Zhang QL, Dong ZX, Xiong Y, et al. Genome-wide transcriptional response of microRNAs to the benzo(a)pyrene stress in amphioxus Branchiostoma belcheri. Chemosphere 2019; 218: 205-10.
[http://dx.doi.org/10.1016/j.chemosphere.2018.11.119] [PMID: 30471501]
[24]
Caiment F, Gaj S, Claessen S, Kleinjans J. High-throughput data integration of RNA-miRNA-circRNA reveals novel insights into mechanisms of benzo[a]pyrene-induced carcinogenicity. Nucleic Acids Res 2015; 43(5): 2525-34.
[http://dx.doi.org/10.1093/nar/gkv115] [PMID: 25690898]
[25]
Deng Q, Huang S, Zhang X, et al. Plasma microRNA expression and micronuclei frequency in workers exposed to polycyclic aromatic hydrocarbons. Environ Health Perspect 2014; 122(7): 719-25.
[http://dx.doi.org/10.1289/ehp.1307080] [PMID: 24633190]
[26]
Halappanavar S, Wu D, Williams A, et al. Pulmonary gene and microRNA expression changes in mice exposed to benzo(a)pyrene by oral gavage. Toxicology 2011; 285(3): 133-41.
[http://dx.doi.org/10.1016/j.tox.2011.04.011] [PMID: 21569818]
[27]
Kuc C, Richard DJ, Johnson S, et al. Rainbow trout exposed to benzo[a]pyrene yields conserved microRNA binding sites in DNA methyltransferases across 500 million years of evolution. Sci Rep 2017; 7(1): 16843.
[http://dx.doi.org/10.1038/s41598-017-17236-x] [PMID: 29203905]
[28]
Li D, Wang Q, Liu C, et al. Aberrant expression of miR-638 contributes to benzo(a)pyrene-induced human cell transformation. Toxicol Sci 2012; 125(2): 382-91.
[http://dx.doi.org/10.1093/toxsci/kfr299] [PMID: 22048643]
[29]
Marrone AK, Tryndyak V, Beland FA, Pogribny IP. MicroRNA responses to the genotoxic carcinogens aflatoxin B1and benzo[a]pyrene in human hepaRG cells. Toxicol Sci 2015; 149(2): 496-502.
[http://dx.doi.org/10.1093/toxsci/kfv253]
[30]
Rinsky RA. Benzene and leukemia: An epidemiologic risk assessment. Environ Health Perspect 1989; 82: 189-91.
[http://dx.doi.org/10.1289/ehp.8982189] [PMID: 2792040]
[31]
Abernethy DJ, Kleymenova EV, Rose J, Recio L, Faiola B. Human CD34+ hematopoietic progenitor cells are sensitive targets for toxicity induced by 1,4-benzoquinone. Toxicol Sci 2004; 79(1): 82-9.
[http://dx.doi.org/10.1093/toxsci/kfh095] [PMID: 14976336]
[32]
Rappaport SM, Kim S, Lan Q, et al. Human benzene metabolism following occupational and environmental exposures. Chem Biol Interact 2010; 184(1-2): 189-95.
[http://dx.doi.org/10.1016/j.cbi.2009.12.017] [PMID: 20026321]
[33]
Liang B, Chen Y, Yuan W, et al. Down-regulation of miRNA-451a and miRNA-486-5p involved in benzene-induced inhibition on erythroid cell differentiation in vitro and in vivo. Arch Toxicol 2018; 92(1): 259-72.
[http://dx.doi.org/10.1007/s00204-017-2033-7] [PMID: 28733890]
[34]
Wei H, Zhang J, Tan K, Sun R, Yin L, Pu Y. Benzene-induced aberrant miRNA expression profile in hematopoietic progenitor cells in C57BL/6 mice. Int J Mol Sci 2015; 16(11): 27058-71.
[http://dx.doi.org/10.3390/ijms161126001] [PMID: 26569237]
[35]
Bai W, Chen Y, Yang J, Niu P, Tian L, Gao A. Aberrant miRNA profiles associated with chronic benzene poisoning. Exp Mol Pathol 2014; 96(3): 426-30.
[http://dx.doi.org/10.1016/j.yexmp.2014.04.011] [PMID: 24780745]
[36]
Chen Y, Sun P, Guo X, Gao A. MiR-34a, a promising novel biomarker for benzene toxicity, is involved in cell apoptosis triggered by 1,4-benzoquinone through targeting Bcl-2. Environ Pollut 2017; 221: 256-65.
[http://dx.doi.org/10.1016/j.envpol.2016.11.072] [PMID: 27939626]
[37]
Liu Y, Chen X, Bian Q, et al. Analysis of plasma microRNA expression profiles in a Chinese population occupationally exposed to benzene and in a population with chronic benzene poisoning. J Thorac Dis 2016; 8(3): 403-14.
[http://dx.doi.org/10.21037/jtd.2016.02.56] [PMID: 27076935]
[38]
Hu D, Peng X, Liu Y, et al. Overexpression of miR-221 in peripheral blood lymphocytes in petrol station attendants: A population based cross-sectional study in southern China. Chemosphere 2016; 149: 8-13.
[http://dx.doi.org/10.1016/j.chemosphere.2016.01.083] [PMID: 26841344]
[39]
Wang F, Li C, Liu W, Jin Y. Modulation of microRNA expression by volatile organic compounds in mouse lung. Environ Toxicol 2014; 29(6): 679-89.
[http://dx.doi.org/10.1002/tox.21795] [PMID: 24733833]
[40]
Piao F, Chen Y, Yu L, et al. 2,5-Hexanedione-induced deregulation of axon-related microRNA expression in rat nerve tissues. Toxicol Lett 2020; 320: 95-102.
[http://dx.doi.org/10.1016/j.toxlet.2019.11.019] [PMID: 31760062]
[41]
Lim JH, Song M-K, Cho Y, Kim W, Han SO, Ryu J-C. Comparative analysis of microRNA and mRNA expression profiles in cells and exosomes under toluene exposure. Toxicol In Vitro 2017; 41: 92-101.
[http://dx.doi.org/10.1016/j.tiv.2017.02.020] [PMID: 28245982]
[42]
Song MK, Park YK, Ryu JC. Polycyclic Aromatic Hydrocarbon (PAH)-mediated upregulation of hepatic microRNA-181 family promotes cancer cell migration by targeting MAPK phosphatase-5, regulating the activation of p38 MAPK. Toxicol Appl Pharmacol 2013; 273(1): 130-9.
[http://dx.doi.org/10.1016/j.taap.2013.08.016] [PMID: 23993976]
[43]
Huang L, Xi Z, Wang C, et al. Phenanthrene exposure induces cardiac hypertrophy via reducing miR-133a expression by DNA methylation. Sci Rep 2016; 6(1): 20105.
[http://dx.doi.org/10.1038/srep20105] [PMID: 26830171]
[44]
Xu N, He D, Shao Y, et al. Lung-derived exosomes in phosgene-induced acute lung injury regulate the functions of mesenchymal stem cells partially via miR-28-5p. Biomed Pharmacother 2020; 121: 109603.
[http://dx.doi.org/10.1016/j.biopha.2019.109603] [PMID: 31707339]
[45]
Wnuk A, Rzemieniec J, Staroń J, et al. Prenatal exposure to benzophenone-3 impairs autophagy, disrupts RXRs/PPARγ signaling, and alters epigenetic and post-translational statuses in brain neurons. Mol Neurobiol 2019; 56(7): 4820-37.
[http://dx.doi.org/10.1007/s12035-018-1401-5] [PMID: 30402708]
[46]
Szabo C. A timeline of hydrogen sulfide (H2S) research: From environmental toxin to biological mediator. Biochem Pharmacol 2018; 149: 5-19.
[http://dx.doi.org/10.1016/j.bcp.2017.09.010] [PMID: 28947277]
[47]
Rumbeiha W, Whitley E, Anantharam P, Kim DS, Kanthasamy A. Acute hydrogen sulfide-induced neuropathology and neurological sequelae: Challenges for translational neuroprotective research. Ann N Y Acad Sci 2016; 1378(1): 5-16.
[http://dx.doi.org/10.1111/nyas.13148] [PMID: 27442775]
[48]
Malone Rubright SL, Pearce LL, Peterson J. Environmental toxicology of hydrogen sulfide. Nitric Oxide 2017; 71: 1-13.
[http://dx.doi.org/10.1016/j.niox.2017.09.011] [PMID: 29017846]
[49]
Yin K, Cui Y, Qu Y, Zhang J, Zhang H, Lin H. Hydrogen sulfide upregulates miR-16-5p targeting PiK3R1 and RAF1 to inhibit neutrophil extracellular trap formation in chickens. Ecotoxicol Environ Saf 2020; 194: 110412.
[http://dx.doi.org/10.1016/j.ecoenv.2020.110412] [PMID: 32155482]
[50]
Middlebrook AM, Murphy DM, Ahmadov R, et al. Air quality implications of the deepwater horizon oil spill. Proc Natl Acad Sci USA 2012; 109(50): 20280-5.
[http://dx.doi.org/10.1073/pnas.1110052108] [PMID: 22205764]
[51]
Chen J, Zhang S, Tong J, et al. Whole transcriptome-based miRNA-mRNA network analysis revealed the mechanism of inflammation-immunosuppressive damage caused by cadmium in common carp spleens. Sci Total Environ 2020; 717: 137081.
[http://dx.doi.org/10.1016/j.scitotenv.2020.137081] [PMID: 32070891]
[52]
Chen M, Li X, Fan R, et al. Cadmium induces BNIP3-dependent autophagy in chicken spleen by modulating miR-33-AMPK axis. Chemosphere 2018; 194: 396-402.
[http://dx.doi.org/10.1016/j.chemosphere.2017.12.026] [PMID: 29223809]
[53]
Chen S, McKinney GJ, Nichols KM, Colbourne JK, Sepúlveda MS. Novel cadmium responsive microRNAs in Daphnia pulex. Environ Sci Technol 2015; 49(24): 14605-13.
[http://dx.doi.org/10.1021/acs.est.5b03988] [PMID: 26550707]
[54]
Chen S, Nichols KM, Poynton HC, Sepúlveda MS. MicroRNAs are involved in cadmium tolerance in Daphnia pulex. Aquat Toxicol 2016; 175: 241-8.
[http://dx.doi.org/10.1016/j.aquatox.2016.03.023] [PMID: 27078211]
[55]
Deng Q, Dai X, Feng W, et al. Co-exposure to metals and polycyclic aromatic hydrocarbons, microRNA expression, and early health damage in coke oven workers. Environ Int 2019; 122: 369-80.
[http://dx.doi.org/10.1016/j.envint.2018.11.056] [PMID: 30503314]
[56]
Fay MJ, Alt LAC, Ryba D, et al. Cadmium nephrotoxicity is associated with altered MicroRNA expression in the rat renal cortex. Toxics 2018; 6(1): 16.
[http://dx.doi.org/10.3390/toxics6010016] [PMID: 29543730]
[57]
Lemaire J, Van der Hauwaert C, Savary G, et al. Cadmium-induced renal cell toxicity is associated with microRNA deregulation. Int J Toxicol 2020; 39(2): 103-14.
[http://dx.doi.org/10.1177/1091581819899039] [PMID: 31934807]
[58]
Li Q, Kappil MA, Li A, et al. Exploring the associations between microRNA expression profiles and environmental pollutants in human placenta from the National Children’s Study (NCS). Epigenetics 2015; 10(9): 793-802.
[http://dx.doi.org/10.1080/15592294.2015.1066960] [PMID: 26252056]
[59]
Tanwar VS, Zhang X, Jagannathan L, Jose CC, Cuddapah S. Cadmium exposure upregulates SNAIL through miR-30 repression in human lung epithelial cells. Toxicol Appl Pharmacol 2019; 373: 1-9.
[http://dx.doi.org/10.1016/j.taap.2019.04.011] [PMID: 30998937]
[60]
Xu P, Guo H, Wang H, et al. Identification and profiling of microRNAs responsive to cadmium toxicity in hepatopancreas of the freshwater crab Sinopotamon henanense. Hereditas 2019; 156(1): 34.
[http://dx.doi.org/10.1186/s41065-019-0110-z] [PMID: 31708719]
[61]
Yuan W, Liu L, Liang L, et al. MiR-122-5p and miR-326-3p: Potential novel biomarkers for early detection of cadmium exposure. Gene 2020; 724: 144156.
[http://dx.doi.org/10.1016/j.gene.2019.144156] [PMID: 31626960]
[62]
Cárdenas-González M, Osorio-Yáñez C, Gaspar-Ramírez O, et al. Environmental exposure to arsenic and chromium in children is associated with kidney injury molecule-1. Environ Res 2016; 150: 653-62.
[http://dx.doi.org/10.1016/j.envres.2016.06.032] [PMID: 27431456]
[63]
Chandra S, Pandey A, Chowdhuri DK. MiRNA profiling provides insights on adverse effects of Cr(VI) in the midgut tissues of Drosophila melanogaster. J Hazard Mater 2015; 283: 558-67.
[http://dx.doi.org/10.1016/j.jhazmat.2014.09.054] [PMID: 25464296]
[64]
Li Y, Li P, Yu S, Zhang J, Wang T, Jia G. MiR-3940-5p associated with genetic damage in workers exposed to hexavalent chromium. Toxicol Lett 2014; 229(1): 319-26.
[http://dx.doi.org/10.1016/j.toxlet.2014.06.033] [PMID: 24973494]
[65]
Pratheeshkumar P, Son YO, Divya SP, et al. Hexavalent chromium induces malignant transformation of human lung bronchial epithelial cells via ROS-dependent activation of miR-21-PDCD4 signaling. Oncotarget 2016; 7(32): 51193-210.
[http://dx.doi.org/10.18632/oncotarget.9967] [PMID: 27323401]
[66]
An J, Cai T, Che H, et al. The changes of miRNA expression in rat hippocampus following chronic lead exposure. Toxicol Lett 2014; 229(1): 158-66.
[http://dx.doi.org/10.1016/j.toxlet.2014.06.002] [PMID: 24960059]
[67]
Dash M, Eid A, Subaiea G, et al. Developmental exposure to lead (Pb) alters the expression of the human tau gene and its products in a transgenic animal model. Neurotoxicology 2016; 55: 154-9.
[http://dx.doi.org/10.1016/j.neuro.2016.06.001] [PMID: 27293183]
[68]
Kong APS, Xiao K, Choi KC, et al. Associations between microRNA (miR-21, 126, 155 and 221), albuminuria and heavy metals in Hong Kong Chinese adolescents. Clin Chim Acta 2012; 413(13-14): 1053-7.
[http://dx.doi.org/10.1016/j.cca.2012.02.014] [PMID: 22405870]
[69]
Liu G, Tian J, Yin H, Yin J, Tang Y. Self-protective transcriptional alterations in ZF4 cells exposed to Pb(NO3 )2 and AgNO3. J Biochem Mol Toxicol 2019; 33(12): e22408.
[http://dx.doi.org/10.1002/jbt.22408] [PMID: 31617658]
[70]
Masoud AM, Bihaqi SW, Alansi B, et al. Altered microRNA, mRNA, and protein expression of neurodegeneration-related biomarkers and their transcriptional and epigenetic modifiers in a human tau transgenic mouse model in response to developmental lead exposure. J Alzheimers Dis 2018; 63(1): 273-82.
[http://dx.doi.org/10.3233/JAD-170824] [PMID: 29614648]
[71]
Sanders AP, Burris HH, Just AC, et al. Altered miRNA expression in the cervix during pregnancy associated with lead and mercury exposure. Epigenomics 2015; 7(6): 885-96.
[http://dx.doi.org/10.2217/epi.15.54] [PMID: 26418635]
[72]
Su P, Zhao F, Cao Z, Zhang J, Aschner M, Luo W. Mir-203-mediated tricellulin mediates lead-induced in vitro loss of blood-cerebrospinal fluid barrier (BCB) function. Toxicol In Vitro 2015; 29(5): 1185-94.
[http://dx.doi.org/10.1016/j.tiv.2015.05.002] [PMID: 25975750]
[73]
Xu M, Yu Z, Hu F, et al. Identification of differential plasma miRNA profiles in Chinese workers with occupational lead exposure. Biosci Rep 2017; 37(5): BSR20171111.
[http://dx.doi.org/10.1042/BSR20171111] [PMID: 28916729]
[74]
Xue C, Kang B, Su P, et al. MicroRNA-106b-5p participates in lead (Pb2+)-induced cell viability inhibition by targeting XIAP in HT-22 and PC12 cells. Toxicol In Vitro 2020; 66: 104876.
[http://dx.doi.org/10.1016/j.tiv.2020.104876] [PMID: 32344020]
[75]
Ding E, Guo J, Bai Y, et al. MiR-92a and miR-486 are potential diagnostic biomarkers for mercury poisoning and jointly sustain NF-κB activity in mercury toxicity. Sci Rep 2017; 7(1): 15980.
[http://dx.doi.org/10.1038/s41598-017-13230-5] [PMID: 29167424]
[76]
Ding E, Zhao Q, Bai Y, et al. Plasma microRNAs expression profile in female workers occupationally exposed to mercury. J Thorac Dis 2016; 8(5): 833-41.
[http://dx.doi.org/10.21037/jtd.2016.03.36] [PMID: 27162656]
[77]
Wilhelm SM. Estimate of mercury emissions to the atmosphere from petroleum. Environ Sci Technol 2001; 35(24): 4704-10.
[http://dx.doi.org/10.1021/es001804h] [PMID: 11775142]
[78]
Kure EH, Sæbø M, Stangeland AM, et al. Molecular responses to toxicological stressors: profiling microRNAs in wild Atlantic salmon (Salmo salar) exposed to acidic aluminum-rich water. Aquat Toxicol 2013; 138-139: 98-104.
[http://dx.doi.org/10.1016/j.aquatox.2013.04.004] [PMID: 23728355]
[79]
Yun J, Yang H, Li X, et al. Up-regulation of miR-297 mediates aluminum oxide nanoparticle-induced lung inflammation through activation of Notch pathway. Environ Pollut 2020; 259: 113839.
[http://dx.doi.org/10.1016/j.envpol.2019.113839] [PMID: 31918133]
[80]
Ge QD, Tan Y, Luo Y, Wang WJ, Zhang H, Xie C. MiR-132, miR-204 and BDNF-TrkB signaling pathway may be involved in spatial learning and memory impairment of the offspring rats caused by fluorine and aluminum exposure during the embryonic stage and into adulthood. Environ Toxicol Pharmacol 2018; 63: 60-8.
[http://dx.doi.org/10.1016/j.etap.2018.08.011] [PMID: 30172012]
[81]
Ge QD, Xie C, Zhang H, et al. Differential expression of miRNAs in the hippocampi of offspring rats exposed to fluorine combined with aluminum during the embryonic stage and into adulthood. Biol Trace Elem Res 2019; 189(2): 463-77.
[http://dx.doi.org/10.1007/s12011-018-1445-4] [PMID: 30033483]
[82]
Al-Eryani L, Jenkins SF, States VA, Pan J, Malone JC, Rai SN. MiRNA expression profiles of premalignant and malignant arsenic-induced skin lesions. In: PLoS One. 2018; 13: p. (8)0202579.
[http://dx.doi.org/10.1371/journal.pone.0202579]
[83]
Martínez-Pacheco M, Hidalgo-Miranda A, Romero-Córdoba S, Valverde M, Rojas E. MRNA and miRNA expression patterns associated to pathways linked to metal mixture health effects. Gene 2014; 533(2): 508-14.
[http://dx.doi.org/10.1016/j.gene.2013.09.049] [PMID: 24080485]
[84]
Mumtaz F, Albeltagy RS, Diab MSM, Abdel Moneim AE, El-Habit OH. Exposure to arsenite and cadmium induces organotoxicity and miRNAs deregulation in male rats. Environ Sci Pollut Res Int 2020; 27(14): 17184-93.
[http://dx.doi.org/10.1007/s11356-020-08306-1] [PMID: 32152865]
[85]
Chen W, Fu W, Deng Q, et al. Multiple metals exposure and chromosome damage: Exploring the mediation effects of microRNAs and their potentials in lung carcinogenesis. Environ Int 2019; 122: 291-300.
[http://dx.doi.org/10.1016/j.envint.2018.11.020] [PMID: 30455104]
[86]
Cui Q, Yu Z, Purisima EO, Wang E. MicroRNA regulation and interspecific variation of gene expression. Trends Genet 2007; 23(8): 372-5.
[http://dx.doi.org/10.1016/j.tig.2007.04.003] [PMID: 17482307]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy