1
|
Rychlik KA, Illingworth EJ, Sillé FCM. Arsenic and the placenta: A review with emphasis on the immune system. Placenta 2025; 160:73-81. [PMID: 39778256 PMCID: PMC11927389 DOI: 10.1016/j.placenta.2024.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 12/09/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025]
Abstract
Chronic arsenic exposure affects over 140 million people globally. While arsenic easily crosses the placenta, the specific mechanisms impacting placental immune cell populations and fetal health are unclear. Maternal arsenic exposure is epidemiologically linked to increased infection risk, mortality, and cancer susceptibility in offspring, emphasizing the importance of understanding placentally-mediated immune effects. This review explores the potential role of the placenta, a key organ for immune transfer to the developing fetus, in mediating chronic low-dose arsenic exposure effects. Examining three potential pathways-direct contaminant transfer, altered immune transfer from the mother, and indirect impact on fetal immune programming via maternal and placental signaling-the review highlights studies associating maternal arsenic levels with immune-related outcomes, including changes in cord blood T cell populations and increased placental inflammation. Placental gene expression analysis reveals alterations in pathways related to oxidative stress, proteasome activity, and aquaglyceroporin transporter expression. Impact on placental DNA methylation and microRNA regulation as well as on trophoblast dysfunction is discussed, with evidence suggesting inhibited trophoblast migration and placental growth factor expression. The complexity of mixtures, nutrition, and environmental interactions add challenges to investigating the placenta's role in immune programming. Despite inconsistent findings on placental morphology alterations, evidence suggests a potential link between arsenic exposure, placental anomalies, and adverse birth outcomes. Further research is crucial to comprehend the effects of prenatal arsenic exposure on trophoblasts, placental immune cells, and subsequent long-term consequences for fetal immune development and birth outcomes.
Collapse
Affiliation(s)
- Kristal A Rychlik
- Public Health Program, School of Health Professions, University of Mary Hardin-Baylor, USA; Johns Hopkins University Bloomberg School of Public Health, USA
| | - Emily J Illingworth
- Johns Hopkins University Bloomberg School of Public Health, USA; iQ Biosciences, Alameda, CA, USA
| | - Fenna C M Sillé
- Johns Hopkins University Bloomberg School of Public Health, USA.
| |
Collapse
|
2
|
Meier MJ, Harrill J, Johnson K, Thomas RS, Tong W, Rager JE, Yauk CL. Progress in toxicogenomics to protect human health. Nat Rev Genet 2025; 26:105-122. [PMID: 39223311 DOI: 10.1038/s41576-024-00767-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
Toxicogenomics measures molecular features, such as transcripts, proteins, metabolites and epigenomic modifications, to understand and predict the toxicological effects of environmental and pharmaceutical exposures. Transcriptomics has become an integral tool in contemporary toxicology research owing to innovations in gene expression profiling that can provide mechanistic and quantitative information at scale. These data can be used to predict toxicological hazards through the use of transcriptomic biomarkers, network inference analyses, pattern-matching approaches and artificial intelligence. Furthermore, emerging approaches, such as high-throughput dose-response modelling, can leverage toxicogenomic data for human health protection even in the absence of predicting specific hazards. Finally, single-cell transcriptomics and multi-omics provide detailed insights into toxicological mechanisms. Here, we review the progress since the inception of toxicogenomics in applying transcriptomics towards toxicology testing and highlight advances that are transforming risk assessment.
Collapse
Affiliation(s)
- Matthew J Meier
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Joshua Harrill
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC, USA
| | - Kamin Johnson
- Predictive Safety Center, Corteva Agriscience, Indianapolis, IN, USA
| | - Russell S Thomas
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC, USA
| | - Weida Tong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, United States Food and Drug Administration, Jefferson, AR, USA
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Julia E Rager
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- The Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, The University of North Carolina, Chapel Hill, NC, USA
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Carole L Yauk
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
3
|
Karmaus PWF. Application of Single Cell Methods in Immunometabolism and Immunotoxicology. CURRENT OPINION IN TOXICOLOGY 2024; 39:100488. [PMID: 39091379 PMCID: PMC11290472 DOI: 10.1016/j.cotox.2024.100488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Recent developments of novel single-cell analysis techniques have rapidly advanced the fields of immunotoxicology and immunometabolism. Single-cell analyses enable the characterization of immune cells, unraveling heterogeneity, and population dynamics in response to cellular perturbations, including toxicant insults and changes in cellular metabolism. This review provides an overview of current technologies and recent discoveries, illustrating an emerging role of single-cell analyses in the field of immunotoxicology and immunometabolism. Various single-cell techniques, including flow cytometry, mass cytometry, multiplexed imaging, and sequencing, together with their applications to studying immunotoxicology and immunometabolism are discussed. This review emphasizes the potential for single-cell analyses to revolutionize our understanding of immune cell heterogeneity, uncover novel cellular therapeutic targets, and pave the way for novel mechanistic insights.
Collapse
Affiliation(s)
- Peer W F Karmaus
- National Institute of Environmental Health Sciences, US
- Institute for Integrative Toxicology, Michigan State University, US
| |
Collapse
|
4
|
Shu C, Street K, Breton CV, Bastain TM, Wilson ML. A review of single-cell transcriptomics and epigenomics studies in maternal and child health. Epigenomics 2024; 16:775-793. [PMID: 38709139 PMCID: PMC11318716 DOI: 10.1080/17501911.2024.2343276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/11/2024] [Indexed: 05/07/2024] Open
Abstract
Single-cell sequencing technologies enhance our understanding of cellular dynamics throughout pregnancy. We outlined the workflow of single-cell sequencing techniques and reviewed single-cell studies in maternal and child health. We conducted a literature review of single cell studies on maternal and child health using PubMed. We summarized the findings from 16 single-cell atlases of the human and mammalian placenta across gestational stages and 31 single-cell studies on maternal exposures and complications including infection, obesity, diet, gestational diabetes, pre-eclampsia, environmental exposure and preterm birth. Single-cell studies provides insights on novel cell types in placenta and cell type-specific marks associated with maternal exposures and complications.
Collapse
Affiliation(s)
- Chang Shu
- Center for Genetic Epidemiology, Division of Epidemiology & Genetics, Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Kelly Street
- Division of Biostatistics, Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Carrie V Breton
- Division of Environmental Health, Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Theresa M Bastain
- Division of Environmental Health, Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Melissa L Wilson
- Division of Disease Prevention, Policy, & Global Health, Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles,CA USA
| |
Collapse
|
5
|
Armstead BE, Lee CS, Chen Y, Zhao R, Chung CS, Fredericks AM, Monaghan SF, Ayala A. Application of single cell multiomics points to changes in chromatin accessibility near calcitonin receptor like receptor and a possible role for adrenomedullin in the post-shock lung. Front Med (Lausanne) 2023; 10:1003121. [PMID: 37113606 PMCID: PMC10126233 DOI: 10.3389/fmed.2023.1003121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Introduction Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a commonly occurring sequelae of traumatic injury resulting from indirect insults like hypovolemic shock and/or extrapulmonary sepsis. The high lethality rate associated with these pathologies outlines the importance of clarifying the "priming" effects seen in the post-shock lung microenvironment, which are understood to bring about a dysregulated or overt immune response when triggered by a secondary systemic infectious/septic challenge culminating in ALI. In this pilot project, we test the hypothesis that application of a single cell multiomics approach can elucidate novel phenotype specific pathways potentially contributing to shock-induced ALI/ARDS. Methods Hypovolemic shock was induced in C57BL/6 (wild-type), PD-1, PD-L1, or VISTA gene deficient male mice, 8-12 weeks old. Wild-type sham surgeries function as negative controls. A total of 24-h post-shock rodents were sacrificed, their lungs harvested and sectioned, with pools prepared from 2 mice per background, and flash frozen on liquid nitrogen. N = 2 biological replicates (representing 4 mice total) were achieved for all treatment groups across genetic backgrounds. Samples were received by the Boas Center for Genomics and Human Genetics, where single cell multiomics libraries were prepared for RNA/ATAC sequencing. The analysis pipeline Cell Ranger ARC was implemented to attain feature linkage assessments across genes of interest. Results Sham (pre-shock) results suggest high chromatin accessibility around calcitonin receptor like receptor (CALCRL) across cellular phenotypes with 17 and 18 feature links, exhibiting positive correlation with gene expression between biological replicates. Similarity between both sample chromatin profiles/linkage arcs is evident. Post-shock wild-type accessibility is starkly reduced across replicates where the number of feature links drops to 1 and 3, again presenting similar replicate profiles. Samples from shocked gene deficient backgrounds displayed high accessibility and similar profiles to the pre-shock lung microenvironment. Conclusion High pre-shock availability of DNA segments and their positive correlation with CALCRL gene expression suggests an apparent regulatory capacity on transcription. Post-shock gene deficient chromatin profiles presented similar results to that of pre-shock wild-type samples, suggesting an influence on CALCRL accessibility. Key changes illustrated in the pre-ALI context of shock may allow for additional resolution of "priming" and "cellular pre-activation/pre-disposition" processes within the lung microenvironment.
Collapse
Affiliation(s)
- Brandon E. Armstead
- Lifespan-Rhode Island Hospital, Providence, RI, United States
- Division of Surgical Research, Department of Surgery, Brown University, Providence, RI, United States
- Pathobiology Graduate Program, Brown University, Providence, RI, United States
| | - Chung Sunny Lee
- Lifespan-Rhode Island Hospital, Providence, RI, United States
- Division of Surgical Research, Department of Surgery, Brown University, Providence, RI, United States
| | - Yaping Chen
- Lifespan-Rhode Island Hospital, Providence, RI, United States
- Division of Surgical Research, Department of Surgery, Brown University, Providence, RI, United States
| | - Runping Zhao
- Lifespan-Rhode Island Hospital, Providence, RI, United States
- Division of Surgical Research, Department of Surgery, Brown University, Providence, RI, United States
| | - Chun-Shiang Chung
- Lifespan-Rhode Island Hospital, Providence, RI, United States
- Division of Surgical Research, Department of Surgery, Brown University, Providence, RI, United States
| | - Alger M. Fredericks
- Lifespan-Rhode Island Hospital, Providence, RI, United States
- Division of Surgical Research, Department of Surgery, Brown University, Providence, RI, United States
- The Miriam Hospital, Providence, RI, United States
- The Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Sean F. Monaghan
- Lifespan-Rhode Island Hospital, Providence, RI, United States
- Division of Surgical Research, Department of Surgery, Brown University, Providence, RI, United States
- Pathobiology Graduate Program, Brown University, Providence, RI, United States
- The Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Alfred Ayala
- Lifespan-Rhode Island Hospital, Providence, RI, United States
- Division of Surgical Research, Department of Surgery, Brown University, Providence, RI, United States
- Pathobiology Graduate Program, Brown University, Providence, RI, United States
- The Warren Alpert Medical School, Brown University, Providence, RI, United States
| |
Collapse
|
6
|
Wu X, Shen F, Jiang G, Xue G, Philips S, Gardner L, Cunningham G, Bales C, Cantor E, Schneider BP. A non-coding GWAS variant impacts anthracycline-induced cardiotoxic phenotypes in human iPSC-derived cardiomyocytes. Nat Commun 2022; 13:7171. [PMID: 36418322 PMCID: PMC9684507 DOI: 10.1038/s41467-022-34917-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/11/2022] [Indexed: 11/24/2022] Open
Abstract
Anthracyclines, widely used to treat breast cancer, have the potential for cardiotoxicity. We have previously identified and validated a germline single nucleotide polymorphism, rs28714259, associated with an increased risk of anthracycline-induced heart failure. We now provide insights into the mechanism by which rs28714259 might confer increased risk of cardiac damage. Using hiPSC-derived cardiomyocyte cell lines with either intrinsic polymorphism or CRISPR-Cas9-mediated deletion of rs28714259 locus, we demonstrate that glucocorticoid receptor signaling activated by dexamethasone pretreatment prior to doxorubicin exposure preserves cardiomyocyte viability and contractility in cardiomyocytes containing the major allele. Homozygous loss of the rs28714259 major allele diminishes dexamethasone's protective effect. We further demonstrate that the risk allele of rs28714259 disrupts glucocorticoid receptor and rs28714259 binding affinity. Finally, we highlight the activation of genes and pathways involved in cardiac hypertrophy signaling that are blocked by the risk allele, suggesting a decreased adaptive survival response to doxorubicin-related stress.
Collapse
Affiliation(s)
- Xi Wu
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Fei Shen
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Guanglong Jiang
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Gloria Xue
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Santosh Philips
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Laura Gardner
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Geneva Cunningham
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Casey Bales
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Erica Cantor
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Bryan Paul Schneider
- Department of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
7
|
Giles BH, Mann KK. Arsenic as an immunotoxicant. Toxicol Appl Pharmacol 2022; 454:116248. [PMID: 36122737 DOI: 10.1016/j.taap.2022.116248] [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: 07/22/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 10/31/2022]
Abstract
Arsenic is world-wide contaminant to which millions of people are exposed. The health consequences of arsenic exposure are varied, including cancer, cardiometabolic disease, and respiratory disorders. Arsenic is also toxic to the immune system, which may link many of the pathologies associated with arsenic exposure. The immune system can be classified into two interconnected arms: the innate and the adaptive immune responses. Herein, we discuss the effects of arsenic on key cell types within each of these arms, highlighting both in vitro and in vivo responses. These cells include macrophages, neutrophils, dendritic cells, and both B and T lymphocytes. Furthermore, we will explore data from human populations where altered immune status is implicated in disease and identify several data gaps where research is needed to complete our understanding of the immunotoxic effects of arsenic.
Collapse
Affiliation(s)
- Braeden H Giles
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Koren K Mann
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.
| |
Collapse
|
8
|
Bulka CM, Enggasser AE, Fry RC. Epigenetics at the Intersection of COVID-19 Risk and Environmental Chemical Exposures. Curr Environ Health Rep 2022; 9:477-489. [PMID: 35648356 PMCID: PMC9157479 DOI: 10.1007/s40572-022-00353-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW Several environmental contaminants have been implicated as contributors to COVID-19 susceptibility and severity. Immunomodulation and epigenetic regulation have been hypothesized as mediators of this relationship, but the precise underlying molecular mechanisms are not well-characterized. This review examines the evidence for epigenetic modification at the intersection of COVID-19 and environmental chemical exposures. RECENT FINDINGS Numerous environmental contaminants including air pollutants, toxic metal(loid)s, per- and polyfluorinated substances, and endocrine disrupting chemicals are hypothesized to increase susceptibility to the SARS-CoV-2 virus and the risk of severe COVID-19, but few studies currently exist. Drawing on evidence that many environmental chemicals alter the epigenetic regulation of key immunity genes and pathways, we discuss how exposures likely perturb host antiviral responses. Specific mechanisms vary by contaminant but include general immunomodulation as well as regulation of viral entry and recognition, inflammation, and immunologic memory pathways, among others. Associations between environmental contaminants and COVID-19 are likely mediated, in part, by epigenetic regulation of key immune pathways involved in the host response to SARS-CoV-2.
Collapse
Affiliation(s)
- Catherine M Bulka
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adam E Enggasser
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rebecca C Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, 166A Rosenau Hall, CB #7431, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
9
|
Haimbaugh A, Meyer D, Akemann C, Gurdziel K, Baker TR. Comparative Toxicotranscriptomics of Single Cell RNA-Seq and Conventional RNA-Seq in TCDD-Exposed Testicular Tissue. FRONTIERS IN TOXICOLOGY 2022; 4:821116. [PMID: 35615540 PMCID: PMC9126299 DOI: 10.3389/ftox.2022.821116] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/03/2022] [Indexed: 12/18/2022] Open
Abstract
In this report, we compare the outcomes and limitations of two methods of transcriptomic inquiry on adult zebrafish testes exposed to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) during sexual differentiation: conventional or bulk RNA-seq (bulk-seq) and single cell RNA sequencing (scRNA-seq) data. scRNA-seq has emerged as a valuable tool for uncovering cell type-specific transcriptome dynamics which exist in heterogeneous tissue. Our lab previously showed the toxicological value of the scRNA-seq pipeline to characterize the sequelae of TCDD exposure in testes, demonstrating that loss of spermatids and spermatozoa, but not other cell types, contributed to the pathology of infertility in adult male zebrafish exposed during sexual differentiation. To investigate the potential for technical artifacts in scRNA-seq such as cell dissociation effects and reduced transcriptome coverage, we compared bulk-sequenced and scRNA-seq-paired samples from control and TCDD-exposed samples to understand what is gained and lost in scRNA-seq vs bulk-seq, both transcriptomically and toxicologically. We hypothesized that the testes may be sensitive to tissue disruption as they contain multiple cell types under constant division and/or maturation, and that TCDD exposure may mediate the extent of sensitivity. Thus, we sought to understand the extent to which this dissociation impacts the toxicological value of data returned from scRNA-seq. We confirm that the required dissociation of individual cells from intact tissue has a significant impact on gene expression, affecting gene pathways with the potential to confound toxicogenomics studies on exposures if findings are not well-controlled and well-situated in context. Additionally, a common scRNA-seq method using cDNA amplified from the 3' end of mRNA under-detects low-expressing transcripts including transcription factors. We confirm this, and show TCDD-related genes may be overlooked by scRNA-seq, however, this under-detection effect is not mediated by TCDD exposure. Even so, scRNA-seq generally extracted toxicologically relevant information better than the bulk-seq method in the present study. This report aims to inform future experimental design for transcriptomic investigation in the growing field of toxicogenomics by demonstrating the differential information extracted from sequencing cells-despite being from the same tissue and exposure scheme-is influenced by the specific protocol used, with implications for the interpretation of exposure-induced risk.
Collapse
Affiliation(s)
- Alex Haimbaugh
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, United States
| | - Danielle Meyer
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, United States
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, United States
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, United States
| | - Camille Akemann
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, United States
| | - Katherine Gurdziel
- Genome Sciences Core, Office of the Vice President for Research, Wayne State University, Detroit, MI, United States
| | - Tracie R. Baker
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, United States
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, United States
- Department of Environmental and Global Health, University of Florida, Gainesville, FL, United States
| |
Collapse
|
10
|
Li M, Gong J, Gao L, Zou T, Kang J, Xu H. Advanced human developmental toxicity and teratogenicity assessment using human organoid models. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 235:113429. [PMID: 35325609 DOI: 10.1016/j.ecoenv.2022.113429] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/12/2022] [Accepted: 03/16/2022] [Indexed: 06/14/2023]
Abstract
Tremendous progress has been made in the field of toxicology leading to the advance of developmental toxicity assessment. Conventional animal models and in vitro two-dimensional models cannot accurately describe toxic effects and predict actual in vivo responses due to obvious inter-species differences between humans and animals, as well as the lack of a physiologically relevant tissue microenvironment. Human embryonic stem cell (hESC)- and induced pluripotent stem cell (iPSC)-derived three-dimensional organoids are ideal complex and multicellular organotypic models, which are indispensable in recapitulating morphogenesis, cellular interactions, and molecular processes of early human organ development. Recently, human organoids have been used for drug discovery, chemical toxicity and safety in vitro assessment. This review discusses the recent advances in the use of human organoid models, (i.e., brain, retinal, cardiac, liver, kidney, lung, and intestinal organoid models) for developmental toxicity and teratogenicity assessment of distinct tissues/organs following exposure to pharmaceutical compounds, heavy metals, persistent organic pollutants, nanomaterials, and ambient air pollutants. Combining next-generation organoid models with innovative engineering technologies generates novel and powerful tools for developmental toxicity and teratogenicity assessment, and the rapid progress in this field is expected to continue.
Collapse
Affiliation(s)
- Minghui Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Jing Gong
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Lixiong Gao
- Department of Ophthalmology, Third Medical Center of PLA General Hospital, Beijing 100039, China
| | - Ting Zou
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Jiahui Kang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China.
| |
Collapse
|
11
|
Magalhaes MS, Potter HG, Ahlback A, Gentek R. Developmental programming of macrophages by early life adversity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 368:213-259. [PMID: 35636928 DOI: 10.1016/bs.ircmb.2022.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Macrophages are central elements of all organs, where they have a multitude of physiological and pathological functions. The first macrophages are produced during fetal development, and most adult organs retain populations of fetal-derived macrophages that self-maintain without major input of hematopoietic stem cell-derived monocytes. Their developmental origins make macrophages highly susceptible to environmental perturbations experienced in early life, in particular the fetal period. It is now well recognized that such adverse developmental conditions contribute to a wide range of diseases later in life. This chapter explores the notion that macrophages are key targets of environmental adversities during development, and mediators of their long-term impact on health and disease. We first briefly summarize our current understanding of macrophage ontogeny and their biology in tissues and consider potential mechanisms by which environmental stressors may mediate fetal programming. We then review evidence for programming of macrophages by adversities ranging from maternal immune activation and diet to environmental pollutants and toxins, which have disease relevance for different organ systems. Throughout this chapter, we contemplate appropriate experimental strategies to study macrophage programming. We conclude by discussing how our current knowledge of macrophage programming could be conceptualized, and finally highlight open questions in the field and approaches to address them.
Collapse
Affiliation(s)
- Marlene S Magalhaes
- Centre for Inflammation Research & Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Harry G Potter
- Centre for Inflammation Research & Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Anna Ahlback
- Centre for Inflammation Research & Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Rebecca Gentek
- Centre for Inflammation Research & Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
12
|
Signes-Pastor AJ, Martinez-Camblor P, Baker E, Madan J, Guill MF, Karagas MR. Prenatal exposure to arsenic and lung function in children from the New Hampshire Birth Cohort Study. ENVIRONMENT INTERNATIONAL 2021; 155:106673. [PMID: 34091160 PMCID: PMC8353991 DOI: 10.1016/j.envint.2021.106673] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 04/21/2021] [Accepted: 05/25/2021] [Indexed: 05/03/2023]
Abstract
Prenatal arsenic exposure is associated with an increased risk of lung cancer along with multiple non-carcinogenic outcomes, including respiratory diseases in arsenic-contaminated areas. Limited epidemiologic data exist on whether in utero arsenic exposure influences lung development and subsequent respiratory health. We investigated the association between gestational arsenic exposure and childhood lung function in the New Hampshire Birth Cohort Study. Urinary arsenic speciation including inorganic arsenic (iAs), monomethylarsonic acid (MMA), dimethylarsinic acid (DMA) and arsenobetaine was measured in maternal urine samples collected during pregnancy and spirometry was performed in offspring at a median age of 7.4 years. Forced vital capacity (FVC), forced expiratory volume in the first second of exhalation (FEV1), and forced expiratory flow between 25% and 75% of FVC (FEF25-75) standardized z-scores were assessed in linear models as dependent variables with the log2-transformed summation of urinary arsenic species (ΣAs = iAs + MMA + DMA) corrected for specific gravity as an independent variable and with adjustment for maternal smoking status, children's age, sex and height. Among the 358 children in the study, a doubling of ΣAs was associated with a -0.08 (ß) decrease in FVC z-scores (95% confidence interval (CI) from -0.14 to -0.01) and -0.10 (ß) (95% CI from -0.18 to -0.02) decrease in FEV1 z-scores. The inverse association appeared stronger among those mothers with lower secondary methylation index (urinary DMA/MMA), especially among girls. No association was observed for FEF25-75 z-scores. Our results suggest that gestation arsenic exposure at levels relevant to the general US population during the vulnerable period of lung formation may adversely affect lung function in childhood.
Collapse
Affiliation(s)
- Antonio J Signes-Pastor
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, United States
| | - Pablo Martinez-Camblor
- Biomedical Data Science Department, Geisel School of Medicine, Dartmouth College, Lebanon, NH, United States
| | - Emily Baker
- Department of Obstetrics & Gynecology, Division of Maternal Fetal Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Juliette Madan
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, United States; Department of Pediatrics, Geisel School of Medicine, Dartmouth College, Lebanon, NH, United States
| | - Margaret F Guill
- Department of Pediatrics, Geisel School of Medicine, Dartmouth College, Lebanon, NH, United States
| | - Margaret R Karagas
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, United States.
| |
Collapse
|
13
|
Russo M, Humes ST, Figueroa AM, Tagmount A, Zhang P, Loguinov A, Lednicky JA, Sabo-Attwood T, Vulpe CD, Liu B. Organochlorine Pesticide Dieldrin Suppresses Cellular Interferon-Related Antiviral Gene Expression. Toxicol Sci 2021; 182:260-274. [PMID: 34051100 DOI: 10.1093/toxsci/kfab064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Organochlorine pesticides (OCPs) are persistent pollutants linked to diverse adverse health outcomes. Environmental exposure to OCPs has been suggested to negatively impact the immune system but their effects on cellular antiviral responses remain unknown. Transcriptomic analysis of N27 rat dopaminergic neuronal cells unexpectedly detected high level expression of genes in the interferon (IFN)-related antiviral response pathways including the IFN-induced protein with tetratricopeptide repeats 1 and 2 (Ifit1/2) and the MX Dynamin Like GTPases Mx1 and Mx2. Interestingly, treatment of N27 cells with dieldrin markedly downregulated the expression of many of these genes. Dieldrin exterted a similar effect in inhibiting IFIT2 and MX1 gene expression in human SH-SY5Y neuronal cells induced by an RNA viral mimic, polyinosinic: polycytidylic acid (poly I:C) and IFIT2/3 gene expression in human pulmonary epithelial cells exposed to human influenza H1N1 virus. Mechanistically, dieldrin induced a rapid rise in levels of intracellular reactive oxygen species (iROS) and a decrease in intracellular glutathione (GSH) levels in SH-SY5Y cells. Treatment with N-acetylcysteine, an antioxidant and GSH biosynthesis precursor, effectively blocked both dieldrin-induced increases in iROS and its inhibition of poly I:C-induced upregulation of IFIT and MX gene expression, suggesting a role for intracellular oxidative status in dieldrin's modulation of antiviral gene expression. This study demonstrates that dieldrin modulates key genes of the cellular innate immune responses that are normally involved in the host's cellular defense against viral infections. Our findings have potential relevance to understanding the organismal effects of environmentally persistent organochlorine contaminants on the mammalian cellular immune system.
Collapse
Affiliation(s)
- Max Russo
- Department of Pharmacodynamics, University of Florida, College of Pharmacy, Gainesville, Florida 32610, USA
| | - Sara T Humes
- Department of Environmental and Global Health, University of Florida, College of Public Health and Health Professions, Gainesville, Florida 32610, USA
| | - Ariana M Figueroa
- Department of Pharmacodynamics, University of Florida, College of Pharmacy, Gainesville, Florida 32610, USA
| | - Abderrahmane Tagmount
- Department of Physiological Sciences, University of Florida, College of Veterinary Medicine, Gainesville, Florida 32610, USA
| | - Ping Zhang
- Department of Pharmacodynamics, University of Florida, College of Pharmacy, Gainesville, Florida 32610, USA
| | - Alex Loguinov
- Department of Physiological Sciences, University of Florida, College of Veterinary Medicine, Gainesville, Florida 32610, USA
| | - John A Lednicky
- Department of Environmental and Global Health, University of Florida, College of Public Health and Health Professions, Gainesville, Florida 32610, USA
| | - Tara Sabo-Attwood
- Department of Environmental and Global Health, University of Florida, College of Public Health and Health Professions, Gainesville, Florida 32610, USA
| | - Chris D Vulpe
- Department of Physiological Sciences, University of Florida, College of Veterinary Medicine, Gainesville, Florida 32610, USA
| | - Bin Liu
- Department of Pharmacodynamics, University of Florida, College of Pharmacy, Gainesville, Florida 32610, USA
| |
Collapse
|
14
|
Song B, Xiong G, Luo H, Zuo Z, Zhou Z, Chang X. Single-cell RNA sequencing of mouse neural stem cell differentiation reveals adverse effects of cadmium on neurogenesis. Food Chem Toxicol 2021; 148:111936. [PMID: 33387572 DOI: 10.1016/j.fct.2020.111936] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/16/2020] [Accepted: 12/15/2020] [Indexed: 12/21/2022]
Abstract
Cadmium (Cd) is a toxic heavy metal and widely exists in the environment. Extensive studies have revealed that Cd exposure can elicit neurotoxicity and potentially interfere with neurogenesis. However, underlying mechanisms by which Cd exposure affects neurogenesis remain unclear. In this study, we performed single-cell RNA sequencing (scRNA-seq) of the differentiated mixture from neonatal mouse Neural Stem Cells (mNSCs) that were exposed to Cd for 24 h and differentiated for 7 days. Our results showed that Cd exposure led to an increase in the differentiation of NSCs into astrocytes while a decrease into neurons. Besides, Cd induced subtype-specific response and dysregulated cell-to-cell communication. Collectively, our scRNA-seq data suggested that Cd had toxic effects on NSCs differentiation at the single-cell level, which offered insight into the potential molecular mechanism of Cd on neurogenesis. Furthermore, our findings provided a new method for assessing the neurodevelopmental toxicity of environmental pollutants.
Collapse
Affiliation(s)
- Bo Song
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China.
| | - Guiya Xiong
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China.
| | - Huan Luo
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China.
| | - Zhenzi Zuo
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China.
| | - Zhijun Zhou
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China.
| | - Xiuli Chang
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China.
| |
Collapse
|