1
|
Memarian P, Bagher Z, Asghari S, Aleemardani M, Seifalian A. Emergence of graphene as a novel nanomaterial for cardiovascular applications. NANOSCALE 2024; 16:12793-12819. [PMID: 38919053 DOI: 10.1039/d4nr00018h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
Cardiovascular diseases (CDs) are the foremost cause of death worldwide. Several promising therapeutic methods have been developed for this approach, including pharmacological, surgical intervention, cell therapy, or biomaterial implantation since heart tissue is incapable of regenerating and healing on its own. The best treatment for heart failure to date is heart transplantation and invasive surgical intervention, despite their invasiveness, donor limitations, and the possibility of being rejected by the patient's immune system. To address these challenges, research is being conducted on less invasive and efficient methods. Consequently, graphene-based materials (GBMs) have attracted a great deal of interest in the last decade because of their exceptional mechanical, electrical, chemical, antibacterial, and biocompatibility properties. An overview of GBMs' applications in the cardiovascular system has been presented in this article. Following a brief explanation of graphene and its derivatives' properties, the potential of GBMs to improve and restore cardiovascular system function by using them as cardiac tissue engineering, stents, vascular bypass grafts,and heart valve has been discussed.
Collapse
Affiliation(s)
- Paniz Memarian
- Nanotechnology and Regenerative Medicine Commercialization Centre, London BioScience Innovation Centre, London, UK.
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Zohreh Bagher
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Department of Tissue Engineering & Regenerative Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sheida Asghari
- Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Mina Aleemardani
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, The University of Sheffield, Sheffield, S3 7HQ, UK.
- Department of Translational Health Science, Bristol Medical School, University of Bristol, Bristol BS1 3NY, UK.
| | - Alexander Seifalian
- Nanotechnology and Regenerative Medicine Commercialization Centre, London BioScience Innovation Centre, London, UK.
| |
Collapse
|
2
|
Francis AP, Meenakshi DU, Ganapathy S, Devasena T. Evaluating the ameliorative effect of nano bis-demethoxy curcumin analog against extrapulmonary toxicity in rat induced by inhaled multi-walled carbon nanotube. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:46641-46651. [PMID: 37710065 DOI: 10.1007/s11356-023-29749-2] [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: 02/22/2023] [Accepted: 09/03/2023] [Indexed: 09/16/2023]
Abstract
Carbon nanotubes (CNTs) exposure in human beings through inhalation may affect pulmonary organs and extrapulmonary organs including liver, kidney, brain, spleen, etc. The toxic effects developed as the result of CNTs exposure made us to explore the beneficial effect of nano bis-demethoxy curcumin analog (NBDMCA) towards multi-walled carbon nanotubes (MWCNTs)-induced toxicity in extrapulmonary organs. The current study described the ameliorative effect of NBDMCA against the toxic effects developed by inhaled MWCNTs in the extrapulmonary organs. The rats are exposed to the fixed aerosol concentration of 5 mg/m3 maintained in inhalation exposure chambers MWCNTs for 15 days as per OECD guidelines. After the exposure with MWCNTs, the animals were treated with NBDMCA (5 mg/kg body weight) with different dose frequencies, i.e., 2 doses per week for 1, 2, and 4 weeks. After treatment duration, the blood was drawn from retro-orbital vein and subjected to biochemical and cytokine analysis. Further the animals were euthanized, and the sample tissues were collected and performed oxidative stress and histopathology. The study results revealed that the intravenous administration of NBDMCA suppresses the extrapulmonary toxicity induced by MWCNTs, i.e., annulling the clinical changes and oxidative stress in various extrapulmonary organs at low doses of NBDMCA, evidenced its antioxidant efficacy. Moreover, use of increased doses provides better reduction in toxic symptoms with negligible side effects confirming the dose-dependent efficacy of NBDMCA. Overall, we suggested that NBDMCA may materialize into an effective compound for the reduction of MWCNTs-induced toxicity.
Collapse
Affiliation(s)
- Arul Prakash Francis
- Centre of Molecular Medicine and Diagnostics (COMManD), Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600 077, India
| | | | - Selvam Ganapathy
- International Institute of Biotechnology and Toxicology (IBAT), Padappai, 601301, India
| | - Thiyagarajan Devasena
- Centre for Nanoscience and Technology, A.C. Tech Campus, Anna University, Chennai, 600025, India.
| |
Collapse
|
3
|
Cary CM, Fournier SB, Adams S, Wang X, Yurkow EJ, Stapleton PA. Single pulmonary nanopolystyrene exposure in late-stage pregnancy dysregulates maternal and fetal cardiovascular function. Toxicol Sci 2024; 199:149-159. [PMID: 38366927 PMCID: PMC11057520 DOI: 10.1093/toxsci/kfae019] [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] [Indexed: 02/19/2024] Open
Abstract
Large-scale production and waste of plastic materials have resulted in widespread environmental contamination by the breakdown product of bulk plastic materials to micro- and nanoplastics (MNPs). The small size of these particles enables their suspension in the air, making pulmonary exposure inevitable. Previous work has demonstrated that xenobiotic pulmonary exposure to nanoparticles during gestation leads to maternal vascular impairments, as well as cardiovascular dysfunction within the fetus. Few studies have assessed the toxicological consequences of maternal nanoplastic (NP) exposure; therefore, the objective of this study was to assess maternal and fetal health after a single maternal pulmonary exposure to polystyrene NP in late gestation. We hypothesized that this acute exposure would impair maternal and fetal cardiovascular function. Pregnant rats were exposed to nanopolystyrene on gestational day 19 via intratracheal instillation. 24 h later, maternal and fetal health outcomes were evaluated. Cardiovascular function was assessed in dams using vascular myography ex vivo and in fetuses in vivo function was measured via ultrasound. Both fetal and placental weight were reduced after maternal exposure to nanopolystyrene. Increased heart weight and vascular dysfunction in the aorta were evident in exposed dams. Maternal exposure led to vascular dysfunction in the radial artery of the uterus, a resistance vessel that controls blood flow to the fetoplacental compartment. Function of the fetal heart, fetal aorta, and umbilical artery after gestational exposure was dysregulated. Taken together, these data suggest that exposure to NPs negatively impacts maternal and fetal health, highlighting the concern of MNPs exposure on pregnancy and fetal development.
Collapse
Affiliation(s)
- C M Cary
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, USA
| | - S B Fournier
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, New Jersey 08854, USA
| | - S Adams
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, USA
| | - X Wang
- Molecular Imaging Core, Rutgers University, Piscataway, New Jersey 08854, USA
| | - E J Yurkow
- Molecular Imaging Core, Rutgers University, Piscataway, New Jersey 08854, USA
| | - P A Stapleton
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854, USA
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, New Jersey 08854, USA
| |
Collapse
|
4
|
Mariappan V, Srinivasan R, Pratheesh R, Jujjuvarapu MR, Pillai AB. Predictive biomarkers for the early detection and management of heart failure. Heart Fail Rev 2024; 29:331-353. [PMID: 37702877 DOI: 10.1007/s10741-023-10347-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/04/2023] [Indexed: 09/14/2023]
Abstract
Cardiovascular disease (CVD) is a serious public health concern whose incidence has been on a rise and is projected by the World Health Organization to be the leading global cause of mortality by 2030. Heart failure (HF) is a complicated syndrome resulting from various CVDs of heterogeneous etiologies and exhibits varying pathophysiology, including activation of inflammatory signaling cascade, apoptosis, fibrotic pathway, and neuro-humoral system, thereby leading to compromised cardiac function. During this process, several biomolecules involved in the onset and progression of HF are released into circulation. These circulating biomolecules could serve as unique biomarkers for the detection of subclinical changes and can be utilized for monitoring disease severity. Hence, it is imperative to identify these biomarkers to devise an early predictive strategy to stop the deterioration of cardiac function caused by these complex cellular events. Furthermore, measurement of multiple biomarkers allows clinicians to divide HF patients into sub-groups for treatment and management based on early health outcomes. The present article provides a comprehensive overview of current omics platform available for discovering biomarkers for HF management. Some of the existing and novel biomarkers for the early detection of HF with special reference to endothelial biology are also discussed.
Collapse
Affiliation(s)
- Vignesh Mariappan
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607402, India
| | - Rajesh Srinivasan
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607402, India
| | - Ravindran Pratheesh
- Department of Neurosurgery, Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607402, India
| | - Muraliswar Rao Jujjuvarapu
- Radiodiagnosis and Imageology, Aware Gleneagles Global Hospital, LB Nagar, Hyderabad, Telangana, 500035, India
| | - Agieshkumar Balakrishna Pillai
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry, 607402, India.
| |
Collapse
|
5
|
Adams S, Stapleton PA. Nanoparticles at the maternal-fetal interface. Mol Cell Endocrinol 2023; 578:112067. [PMID: 37689342 PMCID: PMC10591848 DOI: 10.1016/j.mce.2023.112067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/25/2023] [Accepted: 09/05/2023] [Indexed: 09/11/2023]
Abstract
The increasing production of intentional and unintentional nanoparticles (NPs) has led to their accumulation in the environment as air and ground pollution. The heterogeneity of these particles primarily relies on the NP physicochemical properties (i.e., chemical composition, size, shape, surface chemistry, etc.). Pregnancy represents a vulnerable life stage for both the woman and the developing fetus. The ubiquitous nature of these NPs creates a concern for developmental fetal exposures. At the maternal-fetal interface lies the placenta, a temporary endocrine organ that facilitates nutrient and waste exchange as well as communication between maternal and fetal tissues. Recent evidence in human and animal models identifies that gestational exposure to NPs results in placental translocation leading to local effects and endocrine disruption. Currently, the mechanisms underlying placental translocation and cellular uptake of NPs in the placenta are poorly understood. The purpose of this review is to assess the current understanding of the physiochemical factors influencing NP translocation, cellular uptake, and endocrine disruption at the maternal-fetal interface within the available literature.
Collapse
Affiliation(s)
- S Adams
- Department of Pharmacology and Toxicology, USA
| | - P A Stapleton
- Department of Pharmacology and Toxicology, USA; Environmental Occupational and Health Sciences Institute, Rutgers University, Piscataway, NJ, 08854, USA.
| |
Collapse
|
6
|
Xue J, Zhang Z, Sun Y, Jin D, Guo L, Li X, Zhao D, Feng X, Qi W, Zhu H. Research Progress and Molecular Mechanisms of Endothelial Cells Inflammation in Vascular-Related Diseases. J Inflamm Res 2023; 16:3593-3617. [PMID: 37641702 PMCID: PMC10460614 DOI: 10.2147/jir.s418166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023] Open
Abstract
Endothelial cells (ECs) are widely distributed inside the vascular network, forming a vital barrier between the bloodstream and the walls of blood vessels. These versatile cells serve myriad functions, including the regulation of vascular tension and the management of hemostasis and thrombosis. Inflammation constitutes a cascade of biological responses incited by biological, chemical, or physical stimuli. While inflammation is inherently a protective mechanism, dysregulated inflammation can precipitate a host of vascular pathologies. ECs play a critical role in the genesis and progression of vascular inflammation, which has been implicated in the etiology of numerous vascular disorders, such as atherosclerosis, cardiovascular diseases, respiratory diseases, diabetes mellitus, and sepsis. Upon activation, ECs secrete potent inflammatory mediators that elicit both innate and adaptive immune reactions, culminating in inflammation. To date, no comprehensive and nuanced account of the research progress concerning ECs and inflammation in vascular-related maladies exists. Consequently, this review endeavors to synthesize the contributions of ECs to inflammatory processes, delineate the molecular signaling pathways involved in regulation, and categorize and consolidate the various models and treatment strategies for vascular-related diseases. It is our aspiration that this review furnishes cogent experimental evidence supporting the established link between endothelial inflammation and vascular-related pathologies, offers a theoretical foundation for clinical investigations, and imparts valuable insights for the development of therapeutic agents targeting these diseases.
Collapse
Affiliation(s)
- Jiaojiao Xue
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Ziwei Zhang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Yuting Sun
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Di Jin
- Department of Nephrology, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Liming Guo
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Xiangyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Daqing Zhao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Xiaochun Feng
- Department of Nephropathy and Rheumatology in Children, Children’s Medical Center, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Wenxiu Qi
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Haoyu Zhu
- Department of Nephropathy and Rheumatology in Children, Children’s Medical Center, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| |
Collapse
|
7
|
Cary C, Stapleton P. Determinants and mechanisms of inorganic nanoparticle translocation across mammalian biological barriers. Arch Toxicol 2023; 97:2111-2131. [PMID: 37303009 PMCID: PMC10540313 DOI: 10.1007/s00204-023-03528-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 05/22/2023] [Indexed: 06/13/2023]
Abstract
Biological barriers protect delicate internal tissues from exposures to and interactions with hazardous materials. Primary anatomical barriers prevent external agents from reaching systemic circulation and include the pulmonary, gastrointestinal, and dermal barriers. Secondary barriers include the blood-brain, blood-testis, and placental barriers. The tissues protected by secondary barriers are particularly sensitive to agents in systemic circulation. Neurons of the brain cannot regenerate and therefore must have limited interaction with cytotoxic agents. In the testis, the delicate process of spermatogenesis requires a specific milieu distinct from the blood. The placenta protects the developing fetus from compounds in the maternal circulation that would impair limb or organ development. Many biological barriers are semi-permeable, allowing only materials or chemicals, with a specific set of properties, that easily pass through or between cells. Nanoparticles (particles less than 100 nm) have recently drawn specific concern due to the possibility of biological barrier translocation and contact with distal tissues. Current evidence suggests that nanoparticles translocate across both primary and secondary barriers. It is known that the physicochemical properties of nanoparticles can affect biological interactions, and it has been shown that nanoparticles can breach primary and some secondary barriers. However, the mechanism by which nanoparticles cross biological barriers has yet to be determined. Therefore, the purpose of this review is to summarize how different nanoparticle physicochemical properties interact with biological barriers and barrier products to govern translocation.
Collapse
Affiliation(s)
- Chelsea Cary
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Phoebe Stapleton
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Road, Piscataway, NJ, 08854, USA.
| |
Collapse
|
8
|
Shah SA, Reagan CE, Bresticker JE, Wolpe AG, Good ME, Macal EH, Billcheck HO, Bradley LA, French BA, Isakson BE, Wolf MJ, Epstein FH. Obesity-Induced Coronary Microvascular Disease Is Prevented by iNOS Deletion and Reversed by iNOS Inhibition. JACC Basic Transl Sci 2023; 8:501-514. [PMID: 37325396 PMCID: PMC10264569 DOI: 10.1016/j.jacbts.2022.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 02/04/2023]
Abstract
Coronary microvascular disease (CMD) caused by obesity and diabetes is major contributor to heart failure with preserved ejection fraction; however, the mechanisms underlying CMD are not well understood. Using cardiac magnetic resonance applied to mice fed a high-fat, high-sucrose diet as a model of CMD, we elucidated the role of inducible nitric oxide synthase (iNOS) and 1400W, an iNOS antagonist, in CMD. Global iNOS deletion prevented CMD along with the associated oxidative stress and diastolic and subclinical systolic dysfunction. The 1400W treatment reversed established CMD and oxidative stress and preserved systolic/diastolic function in mice fed a high-fat, high-sucrose diet. Thus, iNOS may represent a therapeutic target for CMD.
Collapse
Affiliation(s)
- Soham A. Shah
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Claire E. Reagan
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Julia E. Bresticker
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Abigail G. Wolpe
- The Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Miranda E. Good
- The Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Edgar H. Macal
- The Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Helen O. Billcheck
- Department of Cardiovascular Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Leigh A. Bradley
- Department of Cardiovascular Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Brent A. French
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Brant E. Isakson
- The Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Matthew J. Wolf
- The Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
- Department of Cardiovascular Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Frederick H. Epstein
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
- The Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
9
|
Cary CM, Seymore TN, Singh D, Vayas KN, Goedken MJ, Adams S, Polunas M, Sunil VR, Laskin DL, Demokritou P, Stapleton PA. Single inhalation exposure to polyamide micro and nanoplastic particles impairs vascular dilation without generating pulmonary inflammation in virgin female Sprague Dawley rats. Part Fibre Toxicol 2023; 20:16. [PMID: 37088832 PMCID: PMC10122824 DOI: 10.1186/s12989-023-00525-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 04/11/2023] [Indexed: 04/25/2023] Open
Abstract
BACKGROUND Exposure to micro- and nanoplastic particles (MNPs) in humans is being identified in both the indoor and outdoor environment. Detection of these materials in the air has made inhalation exposure to MNPs a major cause for concern. One type of plastic polymer found in indoor and outdoor settings is polyamide, often referred to as nylon. Inhalation of combustion-derived, metallic, and carbonaceous aerosols generate pulmonary inflammation, cardiovascular dysfunction, and systemic inflammation. Additionally, due to the additives present in plastics, MNPs may act as endocrine disruptors. Currently there is limited knowledge on potential health effects caused by polyamide or general MNP inhalation. OBJECTIVE The purpose of this study is to assess the toxicological consequences of a single inhalation exposure of female rats to polyamide MNP during estrus by means of aerosolization of MNP. METHODS Bulk polyamide powder (i.e., nylon) served as a representative MNP. Polyamide aerosolization was characterized using particle sizers, cascade impactors, and aerosol samplers. Multiple-Path Particle Dosimetry (MPPD) modeling was used to evaluate pulmonary deposition of MNPs. Pulmonary inflammation was assessed by bronchoalveolar lavage (BAL) cell content and H&E-stained tissue sections. Mean arterial pressure (MAP), wire myography of the aorta and uterine artery, and pressure myography of the radial artery was used to assess cardiovascular function. Systemic inflammation and endocrine disruption were quantified by measurement of proinflammatory cytokines and reproductive hormones. RESULTS Our aerosolization exposure platform was found to generate particles within the micro- and nano-size ranges (thereby constituting MNPs). Inhaled particles were predicted to deposit in all regions of the lung; no overt pulmonary inflammation was observed. Conversely, increased blood pressure and impaired dilation in the uterine vasculature was noted while aortic vascular reactivity was unaffected. Inhalation of MNPs resulted in systemic inflammation as measured by increased plasma levels of IL-6. Decreased levels of 17β-estradiol were also observed suggesting that MNPs have endocrine disrupting activity. CONCLUSIONS These data demonstrate aerosolization of MNPs in our inhalation exposure platform. Inhaled MNP aerosols were found to alter inflammatory, cardiovascular, and endocrine activity. These novel findings will contribute to a better understanding of inhaled plastic particle toxicity.
Collapse
Affiliation(s)
- Chelsea M Cary
- Department of Pharmacology and Toxicology Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Road, 08854, Piscataway, NJ, USA
| | - Talia N Seymore
- Department of Pharmacology and Toxicology Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Road, 08854, Piscataway, NJ, USA
| | - Dilpreet Singh
- Center for Nanotechnology and Nanotoxicology, Department of Environmental Health, Harvard T. H. Chan School of Public Health, Harvard University, 02115, Boston, MA, USA
- Environmental and Occupational Health Sciences Institute (EOHSI), 08854, Piscataway, NJ, USA
| | - Kinal N Vayas
- Department of Pharmacology and Toxicology Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Road, 08854, Piscataway, NJ, USA
| | - Michael J Goedken
- Research Pathology Services, Rutgers University, 08854, Piscataway, NJ, USA
| | - Samantha Adams
- Department of Pharmacology and Toxicology Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Road, 08854, Piscataway, NJ, USA
| | - Marianne Polunas
- Research Pathology Services, Rutgers University, 08854, Piscataway, NJ, USA
| | - Vasanthi R Sunil
- Department of Pharmacology and Toxicology Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Road, 08854, Piscataway, NJ, USA
| | - Debra L Laskin
- Department of Pharmacology and Toxicology Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Road, 08854, Piscataway, NJ, USA
- Environmental and Occupational Health Sciences Institute (EOHSI), 08854, Piscataway, NJ, USA
| | - Philip Demokritou
- Center for Nanotechnology and Nanotoxicology, Department of Environmental Health, Harvard T. H. Chan School of Public Health, Harvard University, 02115, Boston, MA, USA
- Environmental and Occupational Health Sciences Institute (EOHSI), 08854, Piscataway, NJ, USA
- Department of Environmental and Occupational Health and Justice, Rutgers School of Public Health, Rutgers University, 08854, Piscataway, NJ, USA
| | - Phoebe A Stapleton
- Department of Pharmacology and Toxicology Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Road, 08854, Piscataway, NJ, USA.
- Environmental and Occupational Health Sciences Institute (EOHSI), 08854, Piscataway, NJ, USA.
| |
Collapse
|
10
|
Young TL, Scieszka D, Begay JG, Lucas SN, Herbert G, Zychowski K, Hunter R, Salazar R, Ottens AK, Erdely A, Gu H, Campen MJ. Aging influence on pulmonary and systemic inflammation and neural metabolomics arising from pulmonary multi-walled carbon nanotube exposure in apolipoprotein E-deficient and C57BL/6 female mice. Inhal Toxicol 2023; 35:86-100. [PMID: 35037817 PMCID: PMC10037439 DOI: 10.1080/08958378.2022.2026538] [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: 09/28/2021] [Accepted: 01/03/2022] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Environmental exposures exacerbate age-related pathologies, such as cardiovascular and neurodegenerative diseases. Nanoparticulates, and specifically carbon nanomaterials, are a fast-growing contributor to the category of inhalable pollutants, whose risks to health are only now being unraveled. The current study assessed the exacerbating effect of age on multiwalled-carbon nanotube (MWCNT) exposure in young and old C57BL/6 and ApoE-/- mice. MATERIALS AND METHODS Female C57BL/6 and apolipoprotein E-deficient (ApoE-/-) mice, aged 8 weeks and 15 months, were exposed to 0 or 40 µg MWCNT via oropharyngeal aspiration. Pulmonary inflammation, inflammatory bioactivity of serum, and neurometabolic changes were assessed at 24 h post-exposure. RESULTS Pulmonary neutrophil infiltration was induced by MWCNT in bronchoalveolar lavage fluid in both C57BL/6 and ApoE-/-. Macrophage counts decreased with MWCNT exposure in ApoE-/- mice but were unaffected by exposure in C57BL/6 mice. Older mice appeared to have greater MWCNT-induced total protein in lavage fluid. BALF cytokines and chemokines were elevated with MWCNT exposure, but CCL2, CXCL1, and CXCL10 showed reduced responses to MWCNT in older mice. However, no significant serum inflammatory bioactivity was detected. Cerebellar metabolic changes in response to MWCNT were modest, but age and strain significantly influenced metabolite profiles assessed. ApoE-/- mice and older mice exhibited less robust metabolite changes in response to exposure, suggesting a reduced health reserve. CONCLUSIONS Age influences the pulmonary and neurological responses to short-term MWCNT exposure. However, with only the model of moderate aging (15 months) in this study, the responses appeared modest compared to inhaled toxicant impacts in more advanced aging models.
Collapse
Affiliation(s)
- Tamara L. Young
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - David Scieszka
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Jessica G. Begay
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Selita N. Lucas
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Guy Herbert
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | | | - Russell Hunter
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Raul Salazar
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Andrew K. Ottens
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, Richmond, VA 23298
| | - Aaron Erdely
- Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV 26505
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix, AZ, US 85004
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987
| | - Matthew J. Campen
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| |
Collapse
|
11
|
Tang J, Bu W, Hu W, Zhao Z, Liu L, Luo C, Wang R, Fan S, Yu S, Wu Q, Wang X, Zhao X. Ferroptosis Is Involved in Sex-Specific Small Intestinal Toxicity in the Offspring of Adult Mice Exposed to Polystyrene Nanoplastics during Pregnancy. ACS NANO 2023; 17:2440-2449. [PMID: 36728677 DOI: 10.1021/acsnano.2c09729] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Nanoplastics are common contaminants in the living environment. Thus far, no investigations have focused on small intestinal injury in the offspring of adult mice that were exposed to nanoplastics through the respiratory system during pregnancy. Here, we evaluated potential intestinal injury in the offspring of adult mice that were subjected to maternal 80 nm polystyrene nanoparticle (PS-NP) exposure during gestation. PS-NP exposure significantly reduced the birth weight of female mice compared with male mice. However, the adult body weights of the female and male offspring were substantially greater in the PS-NP-exposed groups. Additionally, we found that exposure to PS-NPs during pregnancy caused histological changes in the small intestines of both female and male offspring. Mechanistic analysis revealed upregulation of reactive oxygen species in the small intestines, as indicated by changes in the levels of superoxide dismutase (SOD) and malondialdehyde (MDA). Furthermore, exposure to PS-NPs led to downregulation of GPx4, FTH1, and FTL protein levels, indicating initiation of ferroptosis. Notably, the changes in mRNA expression levels of GPx4, FTH1, and FTL differed between female and male offspring. Although all phenotypes failed to demonstrate classic dose-dependent effects, the data imply that small intestinal toxicity is greater in female offspring than in male offspring. Our results suggest that PS-NP exposure during pregnancy causes sex-specific small intestinal toxicity, which might contribute to reactive oxygen species activation and subsequent ferroptosis. Overall, this study showed toxic effects in offspring after PS-NP exposure during pregnancy.
Collapse
Affiliation(s)
- Juan Tang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Wenxia Bu
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Wenxuan Hu
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Zixuan Zhao
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Lei Liu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Chao Luo
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Rui Wang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Susu Fan
- Nantong University Analysis & Testing Center, Nantong 226019, China
| | - Shali Yu
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Qiyun Wu
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Xiaoke Wang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Xinyuan Zhao
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| |
Collapse
|
12
|
Persiani E, Cecchettini A, Ceccherini E, Gisone I, Morales MA, Vozzi F. Microplastics: A Matter of the Heart (and Vascular System). Biomedicines 2023; 11:264. [PMID: 36830801 PMCID: PMC9953450 DOI: 10.3390/biomedicines11020264] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
Plastic use dramatically increased over the past few years. Besides obvious benefits, the consequent plastic waste and mismanagement in disposal have caused ecological problems. Plastic abandoned in the environment is prone to segregation, leading to the generation of microplastics (MPs) and nanoplastics (NPs), which can reach aquatic and terrestrial organisms. MPs/NPs in water can access fish's bodies through the gills, triggering an inflammatory response in loco. Furthermore, from the gills, plastic fragments can be transported within the circulatory system altering blood biochemical parameters and hormone levels and leading to compromised immunocompetence and angiogenesis. In addition, it was also possible to observe an unbalanced ROS production, damage in vascular structure, and enhanced thrombosis. MPs/NPs led to cardiotoxicity, pericardial oedema, and impaired heart rate in fish cardiac tissue. MPs/NPs effects on aquatic organisms pose serious health hazards and ecological consequences because they constitute the food chain for humans. Once present in the mammalian body, plastic particles can interact with circulating cells, eliciting an inflammatory response, with genotoxicity and cytotoxicity of immune cells, enhanced haemolysis, and endothelium adhesion. The interaction of MPs/NPs with plasma proteins allows their transport to distant organs, including the heart. As a consequence of plastic fragment internalisation into cardiomyocytes, oxidative stress was increased, and metabolic parameters were altered. In this scenario, myocardial damage, fibrosis and impaired electrophysiological values were observed. In summary, MPs/NPs are an environmental stressor for cardiac function in living organisms, and a risk assessment of their influence on the cardiovascular system certainly merits further analysis.
Collapse
Affiliation(s)
- Elisa Persiani
- CNR Institute of Clinical Physiology, via Giuseppe Moruzzi 1, 56124 Pisa, Italy
| | - Antonella Cecchettini
- Department of Clinical and Experimental Medicine, University of Pisa, via Volta 4, 56124 Pisa, Italy
| | - Elisa Ceccherini
- CNR Institute of Clinical Physiology, via Giuseppe Moruzzi 1, 56124 Pisa, Italy
| | - Ilaria Gisone
- CNR Institute of Clinical Physiology, via Giuseppe Moruzzi 1, 56124 Pisa, Italy
| | | | - Federico Vozzi
- CNR Institute of Clinical Physiology, via Giuseppe Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
13
|
Krajnak K, Russ KA, McKinney W, Waugh S, Zheng W, Kan H, Kashon ML, Cumpston J, Fedan JS. Biological effects of crude oil vapor. IV. Cardiovascular effects. Toxicol Appl Pharmacol 2022; 447:116071. [PMID: 35598716 PMCID: PMC9904414 DOI: 10.1016/j.taap.2022.116071] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 02/09/2023]
Abstract
Workers in the oil and gas extraction industry are at risk of inhaling volatile organic compounds. Epidemiological studies suggest oil vapor inhalation may affect cardiovascular health. Thus, in this hazard identification study we investigated the effects of inhalation of crude oil vapor (COV) on cardiovascular function. Male rats were exposed to air or COV (300 ppm) for 6 h (acute), or 6 h/day × 4 d/wk. × 4 wk. (sub-chronic). The effects of COV inhalation were assessed 1, 28, and 90 d post-exposure. Acute exposure to COV resulted in reductions in mean arterial and diastolic blood pressures 1 and 28 d after exposure, changes in nitrate-nitrite and H2O2 levels, and in the expression of transcripts and proteins that regulate inflammation, vascular remodeling, and the synthesis of nitric oxide (NO) in the heart and kidneys. The sub-chronic exposure resulted in a reduced sensitivity to α1-adrenoreceptor-mediated vasoconstriction in vitro 28 d post-exposure, and a reduction in oxidative stress in the heart. Sub-chronic COV exposure led to alterations in the expression of NO synthases and anti-oxidant enzymes, which regulate inflammation and oxidative stress in the heart and kidneys. There seems to be a balance between changes in the expression of transcripts associated with the generation of reactive oxygen species (ROS) and antioxidant enzymes. The ability of antioxidant enzymes to reduce or inhibit the effects of ROS may allow the cardiovascular system to adapt to acute COV exposures. However, sub-chronic exposures may result in longer-lasting negative health consequences on the cardiovascular system.
Collapse
Affiliation(s)
- Kristine Krajnak
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505, United States of America.
| | - Kristen A Russ
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505, United States of America
| | - Walter McKinney
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505, United States of America
| | - Stacey Waugh
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505, United States of America
| | - Wen Zheng
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505, United States of America
| | - Hong Kan
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505, United States of America
| | - Michael L Kashon
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505, United States of America
| | - Jared Cumpston
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505, United States of America
| | - Jeffrey S Fedan
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505, United States of America
| |
Collapse
|
14
|
Sager TM, Umbright CM, Mustafa GM, Roberts JR, Orandle MS, Cumpston JL, McKinney WG, Boots T, Kashon ML, Joseph P. Pulmonary toxicity and gene expression changes in response to whole-body inhalation exposure to multi-walled carbon nanotubes in rats. Inhal Toxicol 2022; 34:200-218. [PMID: 35648795 PMCID: PMC9885491 DOI: 10.1080/08958378.2022.2081386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Purpose: To investigate the molecular mechanisms underlying the pulmonary toxicity induced by exposure to one form of multi-walled carbon nanotubes (MWCNT-7).Materials and methods: Rats were exposed, by whole-body inhalation, to air or an aerosol containing MWCNT-7 particles at target cumulative doses (concentration x time) ranging from 22.5 to 180 (mg/m3)h over a three-day (6 hours/day) period and toxicity and global gene expression profiles were determined in the lungs.Results: MWCNT-7 particles, associated with alveolar macrophages (AMs), were detected in rat lungs following the exposure. Mild to moderate lung pathological changes consisting of increased cellularity, thickening of the alveolar wall, alveolitis, fibrosis, and granuloma formation were detected. Bronchoalveolar lavage (BAL) toxicity parameters such as lactate dehydrogenase activity, number of AMs and polymorphonuclear leukocytes (PMNs), intracellular oxidant generation by phagocytes, and levels of cytokines were significantly (p < 0.05) increased in response to exposure to MWCNT-7. Global gene expression profiling identified several significantly differentially expressed genes (fold change >1.5 and FDR p value <0.05) in all the MWCNT-7 exposed rats. Bioinformatic analysis of the gene expression data identified significant enrichment of several diseases/biological function categories (for example, cancer, leukocyte migration, inflammatory response, mitosis, and movement of phagocytes) and canonical pathways (for example, kinetochore metaphase signaling pathway, granulocyte and agranulocyte adhesion and diapedesis, acute phase response, and LXR/RXR activation). The alterations in the lung toxicity parameters and gene expression changes exhibited a dose-response to the MWCNT exposure.Conclusions: Taken together, the data provided insights into the molecular mechanisms underlying the pulmonary toxicity induced by inhalation exposure of rats to MWCNT-7.
Collapse
Affiliation(s)
- Tina M. Sager
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - Christina M. Umbright
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - Gul Mehnaz Mustafa
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - Jenny R. Roberts
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - Marlene S. Orandle
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - Jared L. Cumpston
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - Walter G. McKinney
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - Theresa Boots
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - Michael L. Kashon
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - Pius Joseph
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| |
Collapse
|
15
|
Development of coronary dysfunction in adult progeny after maternal engineered nanomaterial inhalation during gestation. Sci Rep 2021; 11:19374. [PMID: 34588535 PMCID: PMC8481306 DOI: 10.1038/s41598-021-98818-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/15/2021] [Indexed: 01/09/2023] Open
Abstract
Maternal exposure to environmental contaminants during pregnancy can profoundly influence the risk of developing cardiovascular disease in adult offspring. Our previous studies have demonstrated impaired cardiovascular health, microvascular reactivity, and cardiac function in fetal and young adult progeny after maternal inhalation of nano-sized titanium dioxide (nano-TiO2) aerosols during gestation. The present study was designed to evaluate the development of cardiovascular and metabolic diseases later in adulthood. Pregnant Sprague–Dawley rats were exposed to nano-TiO2 aerosols (~ 10 mg/m3, 134 nm median diameter) for 4 h per day, 5 days per week, beginning on gestational day (GD) 4 and ending on GD 19. Progeny were delivered in-house. Body weight was recorded weekly after birth. After 47 weeks, the body weight of exposed progeny was 9.4% greater compared with controls. Heart weight, mean arterial pressure, and plasma biomarkers of inflammation, dyslipidemia, and glycemic control were recorded at 3, 9 and 12 months of age, with no significant adaptations. While no clinical risk factors (i.e., hypertension, dyslipidemia, or systemic inflammation) emerged pertaining to the development of cardiovascular disease, we identified impaired endothelium-dependent and -independent arteriolar dysfunction and cardiac morphological alterations consistent with myocardial inflammation, degeneration, and necrosis in exposed progeny at 12 months. In conclusion, maternal inhalation of nano-TiO2 aerosols during gestation may promote the development of coronary disease in adult offspring.
Collapse
|
16
|
Kunovac A, Hathaway QA, Pinti MV, Durr AJ, Taylor AD, Goldsmith WT, Garner KL, Nurkiewicz TR, Hollander JM. Enhanced antioxidant capacity prevents epitranscriptomic and cardiac alterations in adult offspring gestationally-exposed to ENM. Nanotoxicology 2021; 15:812-831. [PMID: 33969789 PMCID: PMC8363568 DOI: 10.1080/17435390.2021.1921299] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 04/01/2021] [Accepted: 04/18/2021] [Indexed: 12/16/2022]
Abstract
Maternal engineered nanomaterial (ENM) exposure during gestation has been associated with negative long-term effects on cardiovascular health in progeny. Here, we evaluate an epitranscriptomic mechanism that contributes to these chronic ramifications and whether overexpression of mitochondrial phospholipid hydroperoxide glutathione peroxidase (mPHGPx) can preserve cardiovascular function and bioenergetics in offspring following gestational nano-titanium dioxide (TiO2) inhalation exposure. Wild-type (WT) and mPHGPx (Tg) dams were exposed to nano-TiO2 aerosols with a mass concentration of 12.01 ± 0.50 mg/m3 starting from gestational day (GD) 5 for 360 mins/day for 6 nonconsecutive days over 8 days. Echocardiography was performed in pregnant dams, adult (11-week old) and fetal (GD 14) progeny. Mitochondrial function and global N6-methyladenosine (m6A) content were assessed in adult progeny. MPHGPx enzymatic function was further evaluated in adult progeny and m6A-RNA immunoprecipitation (RIP) was combined with RT-qPCR to evaluate m6A content in the 3'-UTR. Following gestational ENM exposure, global longitudinal strain (GLS) was 32% lower in WT adult offspring of WT dams, with preservation in WT offspring of Tg dams. MPHGPx activity was significantly reduced in WT offspring (29%) of WT ENM-exposed dams, but preserved in the progeny of Tg dams. M6A-RIP-qPCR for the SEC insertion sequence region of mPHGPx revealed hypermethylation in WT offspring from ENM-exposed WT dams, which was thwarted in the presence of the maternal transgene. Our findings implicate that m6A hypermethylation of mPHGPx may be culpable for diminished antioxidant capacity and resultant mitochondrial and cardiac deficits that persist into adulthood following gestational ENM inhalation exposure.
Collapse
Affiliation(s)
- Amina Kunovac
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| | - Quincy A. Hathaway
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| | - Mark V. Pinti
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
- West Virginia University School of Pharmacy, Morgantown, WV, USA
| | - Andrya J. Durr
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Andrew D. Taylor
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - William T. Goldsmith
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Physiology & Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Krista L. Garner
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Physiology & Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Timothy R. Nurkiewicz
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Physiology & Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - John M. Hollander
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
17
|
Fournier SB, D'Errico JN, Adler DS, Kollontzi S, Goedken MJ, Fabris L, Yurkow EJ, Stapleton PA. Nanopolystyrene translocation and fetal deposition after acute lung exposure during late-stage pregnancy. Part Fibre Toxicol 2020; 17:55. [PMID: 33099312 PMCID: PMC7585297 DOI: 10.1186/s12989-020-00385-9] [Citation(s) in RCA: 224] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/15/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Plastic is everywhere. It is used in food packaging, storage containers, electronics, furniture, clothing, and common single-use disposable items. Microplastic and nanoplastic particulates are formed from bulk fragmentation and disintegration of plastic pollution. Plastic particulates have recently been detected in indoor air and remote atmospheric fallout. Due to their small size, microplastic and nanoplastic particulate in the atmosphere can be inhaled and may pose a risk for human health, specifically in susceptible populations. When inhaled, nanosized particles have been shown to translocate across pulmonary cell barriers to secondary organs, including the placenta. However, the potential for maternal-to-fetal translocation of nanosized-plastic particles and the impact of nanoplastic deposition or accumulation on fetal health remain unknown. In this study we investigated whether nanopolystyrene particles can cross the placental barrier and deposit in fetal tissues after maternal pulmonary exposure. RESULTS Pregnant Sprague Dawley rats were exposed to 20 nm rhodamine-labeled nanopolystyrene beads (2.64 × 1014 particles) via intratracheal instillation on gestational day (GD) 19. Twenty-four hours later on GD 20, maternal and fetal tissues were evaluated using fluorescent optical imaging. Fetal tissues were fixed for particle visualization with hyperspectral microscopy. Using isolated placental perfusion, a known concentration of nanopolystyrene was injected into the uterine artery. Maternal and fetal effluents were collected for 180 min and assessed for polystyrene particle concentration. Twenty-four hours after maternal exposure, fetal and placental weights were significantly lower (7 and 8%, respectively) compared with controls. Nanopolystyrene particles were detected in the maternal lung, heart, and spleen. Polystyrene nanoparticles were also observed in the placenta, fetal liver, lungs, heart, kidney, and brain suggesting maternal lung-to-fetal tissue nanoparticle translocation in late stage pregnancy. CONCLUSION These studies confirm that maternal pulmonary exposure to nanopolystyrene results in the translocation of plastic particles to placental and fetal tissues and renders the fetoplacental unit vulnerable to adverse effects. These data are vital to the understanding of plastic particulate toxicology and the developmental origins of health and disease.
Collapse
Affiliation(s)
- Sara B Fournier
- Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Rd, Piscataway, NJ, 08854, USA
| | - Jeanine N D'Errico
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, 160 Frelinghuysen Rd, Piscataway, NJ, 08854, USA
| | - Derek S Adler
- Molecular Imaging Center, Rutgers University, 41 Gordon Rd, Piscataway, NJ, 08854, USA
| | - Stamatina Kollontzi
- Department of Material Science and Engineering, School of Engineering, Rutgers University, 607 Taylor Rd, Piscataway, NJ, 08854, USA
| | - Michael J Goedken
- Research Pathology Services, Rutgers University, Piscataway, NJ, 08854, USA
| | - Laura Fabris
- Department of Material Science and Engineering, School of Engineering, Rutgers University, 607 Taylor Rd, Piscataway, NJ, 08854, USA
| | - Edward J Yurkow
- Molecular Imaging Center, Rutgers University, 41 Gordon Rd, Piscataway, NJ, 08854, USA
| | - Phoebe A Stapleton
- Environmental and Occupational Health Sciences Institute, Rutgers University, 170 Frelinghuysen Rd, Piscataway, NJ, 08854, USA.
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, 160 Frelinghuysen Rd, Piscataway, NJ, 08854, USA.
| |
Collapse
|
18
|
Wright MD, Buckley AJ, Smith R. Estimates of carbon nanotube deposition in the lung: improving quality and robustness. Inhal Toxicol 2020; 32:282-298. [PMID: 32689844 DOI: 10.1080/08958378.2020.1785594] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Carbon nanotube (CNT) in vivo inhalation studies are increasingly providing estimates of the quantity of material deposited in the lung, generally estimated using standard formulae and pulmonary deposition models. These models have typically been developed and validated using data from studies using sphere-like particles. Given the importance of particle morphology to pulmonary deposition, the appropriateness of such an approach was explored to identify any potential limitations. Aerosolized CNT particles typically form 'fiber-like' and/or 'broadly spherical' agglomerates. A review of currently used deposition models indicates that none have been directly validated against results for CNT, however, models for spherical particles have been extensively validated against a wide range of particle sizes and materials and are thus expected to provide reasonable estimates for most 'broadly spherical' CNT particles, although experimental confirmation of this would be of benefit, especially given their low density. The validation of fiber deposition models is significantly less extensive and, in general, focused on larger particles, e.g. asbestos. This raises concerns about the accuracy of deposition estimates for 'fiber-like' CNT particles and recommendations are made for future research to address this. An appreciation of the uncertainties on CNT deposition estimates is important for their interpretation and thus it is recommended that model sensitivity and uncertainty assessments be undertaken. Issues surrounding the measurement and derivation of model input data are also addressed, including instrument responses and particle density assessment options. Recommendations are also made for aerosol characterization to 'future-proof' CNT inhalation studies regarding advances in deposition modeling and toxicological understanding.
Collapse
Affiliation(s)
- Matthew D Wright
- Centre for Radiation, Chemical and Environmental Hazards (CRCE), Public Health England (PHE), Chilton, UK
| | - Alison J Buckley
- Centre for Radiation, Chemical and Environmental Hazards (CRCE), Public Health England (PHE), Chilton, UK
| | - Rachel Smith
- Centre for Radiation, Chemical and Environmental Hazards (CRCE), Public Health England (PHE), Chilton, UK
| |
Collapse
|
19
|
Cazier H, Malgorn C, Fresneau N, Georgin D, Sallustrau A, Chollet C, Tabet JC, Campidelli S, Pinault M, Mayne M, Taran F, Dive V, Junot C, Fenaille F, Colsch B. Development of a Mass Spectrometry Imaging Method for Detecting and Mapping Graphene Oxide Nanoparticles in Rodent Tissues. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2020; 31:1025-1036. [PMID: 32223237 DOI: 10.1021/jasms.9b00070] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Graphene-based nanoparticles are continuously being developed for biomedical applications, and their use raises concerns about their environmental and biological impact. In the literature, some imaging techniques based on fluorescence and radioimaging have been used to explore their fate in vivo. Here, we report on the use of label-free mass spectrometry and mass spectrometry imaging (MSI) for graphene oxide (GO) and reduced graphene oxide (rGO) analyses in rodent tissues. Thereby, we extend previous work by focusing on practical questions to obtain reliable and meaningful images. Specific radical anionic carbon clusters ranging from C2-• to C9-• were observed for both GO and rGO species, with a base peak at m/z 72 under negative laser desorption ionization mass spectrometry (LDI-MS) conditions. Extension to an LDI-MSI method was then performed, thus enabling the efficient detection of GO nanoparticles in lung tissue sections of previously exposed mice. The possibility of quantifying those nanoparticles on tissue sections has also been investigated. Two different ways of building calibration curves (i.e., GO suspensions spotted on tissue sections, or added to lung tissue homogenates) were evaluated and returned similar results, with linear dynamic concentration ranges over at least 2 orders of magnitude. Moreover, intra- and inter-day precision studies have been assessed, with relative standard deviation below 25% for each concentration point of a calibration curve. In conclusion, our study confirms that LDI-MSI is a relevant approach for biodistribution studies of carbon-based nanoparticles, as quantification can be achieved, provided that nanoparticle suspension and manufacturing are carefully controlled.
Collapse
Affiliation(s)
- Hélène Cazier
- INRAE, Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, 91191 Gif-sur-Yvette, France
| | - Carole Malgorn
- INRAE, Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, 91191 Gif-sur-Yvette, France
| | - Nathalie Fresneau
- INRAE, Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, 91191 Gif-sur-Yvette, France
| | - Dominique Georgin
- INRAE, Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, 91191 Gif-sur-Yvette, France
| | - Antoine Sallustrau
- INRAE, Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, 91191 Gif-sur-Yvette, France
| | - Céline Chollet
- INRAE, Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, 91191 Gif-sur-Yvette, France
| | - Jean-Claude Tabet
- INRAE, Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, 91191 Gif-sur-Yvette, France
| | | | - Mathieu Pinault
- INRAE, Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, 91191 Gif-sur-Yvette, France
| | - Martine Mayne
- INRAE, Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, 91191 Gif-sur-Yvette, France
| | - Frédéric Taran
- INRAE, Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, 91191 Gif-sur-Yvette, France
| | - Vincent Dive
- INRAE, Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, 91191 Gif-sur-Yvette, France
| | - Christophe Junot
- INRAE, Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, 91191 Gif-sur-Yvette, France
| | - François Fenaille
- INRAE, Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, 91191 Gif-sur-Yvette, France
| | - Benoit Colsch
- INRAE, Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, 91191 Gif-sur-Yvette, France
| |
Collapse
|
20
|
Schubauer-Berigan MK, Dahm MM, Toennis CA, Sammons DL, Eye T, Kodali V, Zeidler-Erdely PC, Erdely A. Association of occupational exposures with ex vivo functional immune response in workers handling carbon nanotubes and nanofibers. Nanotoxicology 2020; 14:404-419. [PMID: 32031476 PMCID: PMC7121920 DOI: 10.1080/17435390.2020.1717007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 12/12/2019] [Accepted: 01/11/2020] [Indexed: 12/19/2022]
Abstract
The objective of this study was to evaluate the association between carbon nanotube and nanofiber (CNT/F) exposure and ex vivo responses of whole blood challenged with secondary stimulants, adjusting for potential confounders, in a cross-sectional study of 102 workers. Multi-day exposure was measured by CNT/F structure count (SC) and elemental carbon (EC) air concentrations. Demographic, lifestyle and other occupational covariate data were obtained via questionnaire. Whole blood collected from each participant was incubated for 18 hours with and without two microbial stimulants (lipopolysaccharide/LPS and staphylococcal enterotoxin type B/SEB) using TruCulture technology to evaluate immune cell activity. Following incubation, supernatants were preserved and analyzed for protein concentrations. The stimulant:null response ratio for each individual protein was analyzed using multiple linear regression, followed by principal component (PC) analysis to determine whether patterns of protein response were related to CNT/F exposure. Adjusting for confounders, CNT/F metrics (most strongly, the SC-based) were significantly (p < 0.05) inversely associated with stimulant:null ratios of several individual biomarkers: GM-CSF, IFN-γ, interleukin (IL)-2, IL-4, IL-5, IL-10, IL-17, and IL-23. CNT/F metrics were significantly inversely associated with PC1 (a weighted mean of most biomarkers, explaining 25% of the variance in the protein ratios) and PC2 (a biomarker contrast, explaining 14%). Among other occupational exposures, only solvent exposure was significant (inversely related to PC2). CNT/F exposure metrics were uniquely related to stimulant responses in challenged whole blood, illustrating reduced responsiveness to a secondary stimulus. This approach, if replicated in other exposed populations, may present a relatively sensitive method to evaluate human response to CNT/F or other occupational exposures.
Collapse
Affiliation(s)
- Mary K. Schubauer-Berigan
- National Institute for Occupational Safety and Health (NIOSH) Division of Field Studies and Engineering, Cincinnati, OH, USA
- Current address: International Agency for Research on Cancer, Evidence Synthesis and Classification Section; Lyon, France
| | - Matthew M. Dahm
- National Institute for Occupational Safety and Health (NIOSH) Division of Field Studies and Engineering, Cincinnati, OH, USA
| | | | | | - Tracy Eye
- NIOSH Health Effects Laboratory Division, Morgantown, WV, USA
| | - Vamsi Kodali
- NIOSH Health Effects Laboratory Division, Morgantown, WV, USA
| | | | - Aaron Erdely
- NIOSH Health Effects Laboratory Division, Morgantown, WV, USA
| |
Collapse
|
21
|
Cao Y, Luo Y. Pharmacological and toxicological aspects of carbon nanotubes (CNTs) to vascular system: A review. Toxicol Appl Pharmacol 2019; 385:114801. [PMID: 31678607 DOI: 10.1016/j.taap.2019.114801] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/15/2019] [Accepted: 10/30/2019] [Indexed: 01/12/2023]
Abstract
Carbon nanotubes (CNTs) are novel carbon based nanomaterials (NMs) that could be used in many areas ranging from electronics to biotechnology. The present review summarized pharmacological and toxicological aspects of CNTs to vascular systems, because the vascular systems are important targets for CNTs during manufacturing process, daily contact and biomedical uses. Functionalized CNTs could be used as novel nanoplateforms to regulate angiogenesis for cancer therapy, as well as nanocarriers to cross blood brain barrier (BBB), one of the major obstacles to prevent the entering of therapeutic substances into brains. However, it has also been shown that inhalational or intravenous contact with CNTs might induce adverse vascular effects, such as progression of atherosclerotic plaque, vasomotor dysfunction, and changes of blood pressure and/or heart rate in laboratory animals, although currently there are only limited reports obtained from CNT-exposed human beings and the results are inconclusive. The mechanisms associated with the vascular toxicity of CNTs remain poorly understood, and it appears that multiple signaling pathways are likely to be involved. The toxicity of CNTs to vascular systems might be reduced by controlling the physicochemical properties of CNTs, particularly lengths, diameters and surface chemistry. At present, the beneficial and adverse effects of CNTs to vascular systems are still largely unknown and require further extensive studies.
Collapse
Affiliation(s)
- Yi Cao
- Key Laboratory of Environment-Friendly Chemistry and Application of Ministry of Education, Lab of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan 411105, PR China.
| | - Yingmei Luo
- Key Laboratory of Environment-Friendly Chemistry and Application of Ministry of Education, Lab of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan 411105, PR China
| |
Collapse
|
22
|
Hajipour MJ, Mehrani M, Abbasi SH, Amin A, Kassaian SE, Garbern JC, Caracciolo G, Zanganeh S, Chitsazan M, Aghaverdi H, Shahri SMK, Ashkarran A, Raoufi M, Bauser-Heaton H, Zhang J, Muehlschlegel JD, Moore A, Lee RT, Wu JC, Serpooshan V, Mahmoudi M. Nanoscale Technologies for Prevention and Treatment of Heart Failure: Challenges and Opportunities. Chem Rev 2019; 119:11352-11390. [PMID: 31490059 PMCID: PMC7003249 DOI: 10.1021/acs.chemrev.8b00323] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The adult myocardium has a limited regenerative capacity following heart injury, and the lost cells are primarily replaced by fibrotic scar tissue. Suboptimal efficiency of current clinical therapies to resurrect the infarcted heart results in injured heart enlargement and remodeling to maintain its physiological functions. These remodeling processes ultimately leads to ischemic cardiomyopathy and heart failure (HF). Recent therapeutic approaches (e.g., regenerative and nanomedicine) have shown promise to prevent HF postmyocardial infarction in animal models. However, these preclinical, clinical, and technological advancements have yet to yield substantial enhancements in the survival rate and quality of life of patients with severe ischemic injuries. This could be attributed largely to the considerable gap in knowledge between clinicians and nanobioengineers. Development of highly effective cardiac regenerative therapies requires connecting and coordinating multiple fields, including cardiology, cellular and molecular biology, biochemistry and chemistry, and mechanical and materials sciences, among others. This review is particularly intended to bridge the knowledge gap between cardiologists and regenerative nanomedicine experts. Establishing this multidisciplinary knowledge base may help pave the way for developing novel, safer, and more effective approaches that will enable the medical community to reduce morbidity and mortality in HF patients.
Collapse
Affiliation(s)
| | - Mehdi Mehrani
- Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ahmad Amin
- Rajaie Cardiovascular, Medical and Research Center, Iran University of Medical Science Tehran, Iran
| | | | - Jessica C. Garbern
- Department of Stem Cell and Regenerative Biology, Harvard University, Harvard Stem Cell Institute, Cambridge, Massachusetts, United States
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts, United States
| | - Giulio Caracciolo
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, 00161, Rome, Italy
| | - Steven Zanganeh
- Department of Radiology, Memorial Sloan Kettering, New York, NY 10065, United States
| | - Mitra Chitsazan
- Rajaie Cardiovascular, Medical and Research Center, Iran University of Medical Science Tehran, Iran
| | - Haniyeh Aghaverdi
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Seyed Mehdi Kamali Shahri
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Aliakbar Ashkarran
- Precision Health Program, Michigan State University, East Lansing, MI, United States
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Mohammad Raoufi
- Physical Chemistry I, Department of Chemistry and Biology & Research Center of Micro and Nanochemistry and Engineering, University of Siegen, Siegen, Germany
| | - Holly Bauser-Heaton
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Jianyi Zhang
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Jochen D. Muehlschlegel
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Anna Moore
- Precision Health Program, Michigan State University, East Lansing, MI, United States
| | - Richard T. Lee
- Department of Stem Cell and Regenerative Biology, Harvard University, Harvard Stem Cell Institute, Cambridge, Massachusetts, United States
- Department of Medicine, Division of Cardiology, Brigham and Women’s Hospital and Harvard Medical School, Cambridge, Massachusetts, United States
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, United States
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, California, United States
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, United States
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, United States
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Morteza Mahmoudi
- Precision Health Program, Michigan State University, East Lansing, MI, United States
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Connors Center for Women’s Health & Gender Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
23
|
Fournier SB, Kallontzi S, Fabris L, Love C, Stapleton PA. Effect of Gestational Age on Maternofetal Vascular Function Following Single Maternal Engineered Nanoparticle Exposure. Cardiovasc Toxicol 2019; 19:321-333. [PMID: 30734150 PMCID: PMC6642065 DOI: 10.1007/s12012-019-09505-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Normal pregnancy outcome is accomplished, in part, by rapid and expansive physiological adaptations to the systemic circulation, the extent of which is specific to gestational day (GD) and anatomical location. Pregnancy-related hemodynamic changes in uterine placental blood flow stimulate compensatory vascular signaling and remodeling that begins early and continues throughout gestation. Exposure of the maternal environment to engineered nanomaterials (ENM) during pregnancy has been shown to impact health of the dam, fetus, and adult offspring; however, the consequences of specific temporal (gestational age) and spatial (vascular location) considerations are largely undetermined. We exposed pregnant Sprague-Dawley rats to nano-TiO2 aerosols at three critical periods of fetal development (GD 4, 12, and 17) to identify vascular perturbations associated with ENM exposure at these developmental milestones. Vascular reactivity of the maternal thoracic aorta, the uterine artery, the umbilical vein, and the fetal thoracic aorta were evaluated using wire myography on GD 20. While impairments were noted at each level of the maternofetal vascular tree and at each exposure day, our results indicate the greatest effects may be identified within the fetal vasculature (umbilical vein and fetal aorta), wherein effects of a single maternal inhalational exposure to nano-TiO2 on GD 4 modified responses to cholinergic, NO, and α-adrenergic signaling.
Collapse
Affiliation(s)
- S B Fournier
- Environmental and Occupational Health Sciences Institute, 170 Frelinghuysen Rd, Piscataway, NJ, 08854, USA
| | - S Kallontzi
- Materials Science and Engineering, Rutgers University, 607 Taylor Rd, Piscataway, NJ, 08854, USA
| | - L Fabris
- Materials Science and Engineering, Rutgers University, 607 Taylor Rd, Piscataway, NJ, 08854, USA
| | - C Love
- Biology and Environmental Studies, Grinnell College, 1116 Eighth Ave, Grinnell, IA, 50112, USA
| | - P A Stapleton
- Environmental and Occupational Health Sciences Institute, 170 Frelinghuysen Rd, Piscataway, NJ, 08854, USA.
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, 160 Frelinghuysen Rd, Piscataway, NJ, 08854, USA.
| |
Collapse
|
24
|
Kunovac A, Hathaway QA, Pinti MV, Goldsmith WT, Durr AJ, Fink GK, Nurkiewicz TR, Hollander JM. ROS promote epigenetic remodeling and cardiac dysfunction in offspring following maternal engineered nanomaterial (ENM) exposure. Part Fibre Toxicol 2019; 16:24. [PMID: 31215478 PMCID: PMC6582485 DOI: 10.1186/s12989-019-0310-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 06/06/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Nano-titanium dioxide (nano-TiO2) is amongst the most widely utilized engineered nanomaterials (ENMs). However, little is known regarding the consequences maternal ENM inhalation exposure has on growing progeny during gestation. ENM inhalation exposure has been reported to decrease mitochondrial bioenergetics and cardiac function, though the mechanisms responsible are poorly understood. Reactive oxygen species (ROS) are increased as a result of ENM inhalation exposure, but it is unclear whether they impact fetal reprogramming. The purpose of this study was to determine whether maternal ENM inhalation exposure influences progeny cardiac development and epigenomic remodeling. RESULTS Pregnant FVB dams were exposed to nano-TiO2 aerosols with a mass concentration of 12.09 ± 0.26 mg/m3 starting at gestational day five (GD 5), for 6 h over 6 non-consecutive days. Aerosol size distribution measurements indicated an aerodynamic count median diameter (CMD) of 156 nm with a geometric standard deviation (GSD) of 1.70. Echocardiographic imaging was used to assess cardiac function in maternal, fetal (GD 15), and young adult (11 weeks) animals. Electron transport chain (ETC) complex activities, mitochondrial size, complexity, and respiration were evaluated, along with 5-methylcytosine, Dnmt1 protein expression, and Hif1α activity. Cardiac functional analyses revealed a 43% increase in left ventricular mass and 25% decrease in cardiac output (fetal), with an 18% decrease in fractional shortening (young adult). In fetal pups, hydrogen peroxide (H2O2) levels were significantly increased (~ 10 fold) with a subsequent decrease in expression of the antioxidant enzyme, phospholipid hydroperoxide glutathione peroxidase (GPx4). ETC complex activity IV was decreased by 68 and 46% in fetal and young adult cardiac mitochondria, respectively. DNA methylation was significantly increased in fetal pups following exposure, along with increased Hif1α activity and Dnmt1 protein expression. Mitochondrial ultrastructure, including increased size, was observed at both fetal and young adult stages following maternal exposure. CONCLUSIONS Maternal inhalation exposure to nano-TiO2 results in adverse effects on cardiac function that are associated with increased H2O2 levels and dysregulation of the Hif1α/Dnmt1 regulatory axis in fetal offspring. Our findings suggest a distinct interplay between ROS and epigenetic remodeling that leads to sustained cardiac contractile dysfunction in growing and young adult offspring following maternal ENM inhalation exposure.
Collapse
Affiliation(s)
- Amina Kunovac
- Division of Exercise Physiology, West Virginia University School of Medicine, PO Box 9227, 1 Medical Center Drive, Morgantown, WV 26506 USA
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV USA
| | - Quincy A. Hathaway
- Division of Exercise Physiology, West Virginia University School of Medicine, PO Box 9227, 1 Medical Center Drive, Morgantown, WV 26506 USA
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV USA
| | - Mark V. Pinti
- West Virginia University School of Pharmacy, Morgantown, WV USA
| | - William T. Goldsmith
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV USA
- Department of Physiology, Pharmacology, Morgantown, WV USA
| | - Andrya J. Durr
- Division of Exercise Physiology, West Virginia University School of Medicine, PO Box 9227, 1 Medical Center Drive, Morgantown, WV 26506 USA
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV USA
| | - Garrett K. Fink
- Division of Exercise Physiology, West Virginia University School of Medicine, PO Box 9227, 1 Medical Center Drive, Morgantown, WV 26506 USA
| | - Timothy R. Nurkiewicz
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV USA
- Department of Physiology, Pharmacology, Morgantown, WV USA
| | - John M. Hollander
- Division of Exercise Physiology, West Virginia University School of Medicine, PO Box 9227, 1 Medical Center Drive, Morgantown, WV 26506 USA
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV USA
| |
Collapse
|
25
|
Snyder-Talkington BN, Dong C, Castranova V, Qian Y, Guo NL. Differential gene regulation in human small airway epithelial cells grown in monoculture versus coculture with human microvascular endothelial cells following multiwalled carbon nanotube exposure. Toxicol Rep 2019; 6:482-488. [PMID: 31194188 PMCID: PMC6554470 DOI: 10.1016/j.toxrep.2019.05.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 05/08/2019] [Accepted: 05/27/2019] [Indexed: 12/20/2022] Open
Abstract
Coculture gene expression may have opposite direction of changes than monoculture. Cells grow and treated in monoculture may exaggerate toxicological responses. Coculture of cells may provide a more in-depth assessment of toxicological responses.
Concurrent with rising production of carbon-based engineered nanomaterials is a potential increase in respiratory and cardiovascular diseases due to exposure to nanomaterials in the workplace atmosphere. While single-cell models of pulmonary exposure are often used to determine the potential toxicity of nanomaterials in vitro, previous studies have shown that coculture cell models better represent the cellular response and crosstalk that occurs in vivo. This study identified differential gene regulation in human small airway epithelial cells (SAECs) grown either in monoculture or in coculture with human microvascular endothelial cells following exposure of the SAECs to multiwalled carbon nanotubes (MWCNTs). SAEC genes that either changed their regulation direction from upregulated in monoculture to downregulated in coculture (or vice versa) or had a more than a two-fold changed in the same regulation direction were identified. Genes that changed regulation direction were most often involved in the processes of cellular growth and proliferation and cellular immune response and inflammation. Genes that had a more than a two-fold change in regulation in the same direction were most often involved in the inflammatory response. The direction and fold-change of this differential gene regulation suggests that toxicity testing in monoculture may exaggerate cellular responses to MWCNTs, and coculture of cells may provide a more in-depth assessment of toxicological responses.
Collapse
Affiliation(s)
- Brandi N Snyder-Talkington
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, 26506, United States
| | - Chunlin Dong
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, 26506, United States
| | - Vincent Castranova
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, 26506, United States
| | - Yong Qian
- National Institute for Occupational and Environmental Safety and Health, 1095 Willowdale Rd., Morgantown, WV, 26505, United States
| | - Nancy L Guo
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, 26506, United States.,Department of Occupational and Environmental Health Sciences, School of Public Health, West Virginia University, Morgantown, WV, 26506, United States
| |
Collapse
|
26
|
Airway Exposure to Modified Multi-walled Carbon Nanotubes Perturbs Cardiovascular Adenosinergic Signaling in Mice. Cardiovasc Toxicol 2019; 19:168-177. [PMID: 30382549 DOI: 10.1007/s12012-018-9487-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The broad list of commercial applications for multi-walled carbon nanotubes (MWCNT) can be further expanded with the addition of various surface chemistry modifications. For example, standard commercial grade MWCNT (C-grade) can be carboxylated (COOH) or nitrogen-doped (N-doped) to suite specific utilities. We previously reported dose-dependent expansions of cardiac ischemia/reperfusion (I/R) injury, 24 h after intratracheal instillation of C-grade, COOH, or N-doped MWCNT in mice. Here, we have tested the hypothesis that airway exposure to MWCNT perturbs cardiovascular adenosinergic signaling, which could contribute to exacerbation of cardiac I/R injury. 100 µL of Vehicle or identical suspension volumes containing 100 µg of C-grade, COOH, or N-doped MWCNT were instilled into the trachea of CD-1 ICR mice. 1 day later, we measured cyclic adenosine monophosphate (cAMP) concentrations in cardiac tissue and evaluated arterial adenosinergic smooth muscle signaling mechanisms related to nitric oxide synthase (NOS) and cyclooxygenase (COX) in isolated aortic tissue. We also verified cardiac I/R injury expansion and examined both lung histology and bronchoalveolar lavage fluid cellularity in MWCNT exposed mice. Myocardial cAMP concentrations were reduced (p < 0.05) in the C-grade group by 17.4% and N-doped group by 13.7% compared to the Vehicle group. Curve fits to aortic ring 2-Cl-Adenosine concentration responses were significantly greater in the MWCNT groups vs. the Vehicle group. Aortic constrictor responses were more pronounced with NOS inhibition and were abolished with COX inhibition. These findings indicate that addition of functional chemical moieties on the surface of MWCNT may alter the biological responses to exposure by influencing cardiovascular adenosinergic signaling and promoting cardiac injury.
Collapse
|
27
|
Hathaway QA, Durr AJ, Shepherd DL, Pinti MV, Brandebura AN, Nichols CE, Kunovac A, Goldsmith WT, Friend SA, Abukabda AB, Fink GK, Nurkiewicz TR, Hollander JM. miRNA-378a as a key regulator of cardiovascular health following engineered nanomaterial inhalation exposure. Nanotoxicology 2019; 13:644-663. [PMID: 30704319 PMCID: PMC6629495 DOI: 10.1080/17435390.2019.1570372] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/13/2018] [Accepted: 12/12/2018] [Indexed: 12/31/2022]
Abstract
Nano-titanium dioxide (nano-TiO2), though one of the most utilized and produced engineered nanomaterials (ENMs), diminishes cardiovascular function through dysregulation of metabolism and mitochondrial bioenergetics following inhalation exposure. The molecular mechanisms governing this cardiac dysfunction remain largely unknown. The purpose of this study was to elucidate molecular mediators that connect nano-TiO2 exposure with impaired cardiac function. Specifically, we were interested in the role of microRNA (miRNA) expression in the resulting dysfunction. Not only are miRNA global regulators of gene expression, but also miRNA-based therapeutics provide a realistic treatment modality. Wild type and MiRNA-378a knockout mice were exposed to nano-TiO2 with an aerodynamic diameter of 182 ± 1.70 nm and a mass concentration of 11.09 mg/m3 for 4 h. Cardiac function, utilizing the Vevo 2100 Imaging System, electron transport chain complex activities, and mitochondrial respiration assessed cardiac and mitochondrial function. Immunoblotting and qPCR examined molecular targets of miRNA-378a. MiRNA-378a-3p expression was increased 48 h post inhalation exposure to nano-TiO2. Knockout of miRNA-378a preserved cardiac function following exposure as revealed by preserved E/A ratio and E/SR ratio. In knockout animals, complex I, III, and IV activities (∼2- to 6-fold) and fatty acid respiration (∼5-fold) were significantly increased. MiRNA-378a regulated proteins involved in mitochondrial fusion, transcription, and fatty acid metabolism. MiRNA-378a-3p acts as a negative regulator of mitochondrial metabolic and biogenesis pathways. MiRNA-378a knockout animals provide a protective effect against nano-TiO2 inhalation exposure by altering mitochondrial structure and function. This is the first study to manipulate a miRNA to attenuate the effects of ENM exposure.
Collapse
Affiliation(s)
- Quincy A. Hathaway
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
- Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Andrya J. Durr
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Danielle L. Shepherd
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Mark V. Pinti
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Ashley N. Brandebura
- Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Cody E. Nichols
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Amina Kunovac
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - William T. Goldsmith
- Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Physiology, Pharmacology & Neuroscience, Morgantown, WV, USA
| | - Sherri A. Friend
- National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Alaeddin B. Abukabda
- Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Physiology, Pharmacology & Neuroscience, Morgantown, WV, USA
| | - Garrett K. Fink
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Timothy R. Nurkiewicz
- Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Physiology, Pharmacology & Neuroscience, Morgantown, WV, USA
| | - John M. Hollander
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
28
|
Snyder-Talkington BN, Dong C, Singh S, Raese R, Qian Y, Porter DW, Wolfarth MG, Guo NL. Multi-Walled Carbon Nanotube-Induced Gene Expression Biomarkers for Medical and Occupational Surveillance. Int J Mol Sci 2019; 20:E2635. [PMID: 31146342 PMCID: PMC6600433 DOI: 10.3390/ijms20112635] [Citation(s) in RCA: 10] [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] [Received: 05/01/2019] [Revised: 05/21/2019] [Accepted: 05/24/2019] [Indexed: 12/19/2022] Open
Abstract
As the demand for multi-walled carbon nanotube (MWCNT) incorporation into industrial and biomedical applications increases, so does the potential for unintentional pulmonary MWCNT exposure, particularly among workers during manufacturing. Pulmonary exposure to MWCNTs raises the potential for development of lung inflammation, fibrosis, and cancer among those exposed; however, there are currently no effective biomarkers for detecting lung fibrosis or predicting the risk of lung cancer resulting from MWCNT exposure. To uncover potential mRNAs and miRNAs that could be used as markers of exposure, this study compared in vivo mRNA and miRNA expression in lung tissue and blood of mice exposed to MWCNTs with in vitro mRNA and miRNA expression from a co-culture model of human lung epithelial and microvascular cells, a system previously shown to have a higher overall genome-scale correlation with mRNA expression in mouse lungs than either cell type grown separately. Concordant mRNAs and miRNAs identified by this study could be used to drive future studies confirming human biomarkers of MWCNT exposure. These potential biomarkers could be used to assess overall worker health and predict the occurrence of MWCNT-induced diseases.
Collapse
Affiliation(s)
| | - Chunlin Dong
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA.
| | - Salvi Singh
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA.
| | - Rebecca Raese
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA.
| | - Yong Qian
- National Institute for Occupational and Environmental Safety and Health, 1095 Willowdale Rd., Morgantown, WV 26505, USA.
| | - Dale W Porter
- National Institute for Occupational and Environmental Safety and Health, 1095 Willowdale Rd., Morgantown, WV 26505, USA.
| | - Michael G Wolfarth
- National Institute for Occupational and Environmental Safety and Health, 1095 Willowdale Rd., Morgantown, WV 26505, USA.
| | - Nancy L Guo
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA.
- Department of Occupational and Environmental Health Sciences, School of Public Health, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
29
|
Mostovenko E, Young T, Muldoon PP, Bishop L, Canal CG, Vucetic A, Zeidler-Erdely PC, Erdely A, Campen MJ, Ottens AK. Nanoparticle exposure driven circulating bioactive peptidome causes systemic inflammation and vascular dysfunction. Part Fibre Toxicol 2019; 16:20. [PMID: 31142334 PMCID: PMC6542040 DOI: 10.1186/s12989-019-0304-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 05/10/2019] [Indexed: 12/22/2022] Open
Abstract
Background The mechanisms driving systemic effects consequent pulmonary nanoparticle exposure remain unclear. Recent work has established the existence of an indirect process by which factors released from the lung into the circulation promote systemic inflammation and cellular dysfunction, particularly on the vasculature. However, the composition of circulating contributing factors and how they are produced remains unknown. Evidence suggests matrix protease involvement; thus, here we used a well-characterized multi-walled carbon nanotube (MWCNT) oropharyngeal aspiration model with known vascular effects to assess the distinct contribution of nanoparticle-induced peptide fragments in driving systemic pathobiology. Results Data-independent mass spectrometry enabled the unbiased quantitative characterization of 841 significant MWCNT-responses within an enriched peptide fraction, with 567 of these factors demonstrating significant correlation across animal-paired bronchoalveolar lavage and serum biofluids. A database search curated for known matrix protease substrates and predicted signaling motifs enabled identification of 73 MWCNT-responsive peptides, which were significantly associated with an abnormal cardiovascular phenotype, extracellular matrix organization, immune-inflammatory processes, cell receptor signaling, and a MWCNT-altered serum exosome population. Production of a diverse peptidomic response was supported by a wide number of upregulated matrix and lysosomal proteases in the lung after MWCNT exposure. The peptide fraction was then found bioactive, producing endothelial cell inflammation and vascular dysfunction ex vivo akin to that induced with whole serum. Results implicate receptor ligand functionality in driving systemic effects, exemplified by an identified 59-mer thrombospondin fragment, replete with CD36 modulatory motifs, that when synthesized produced an anti-angiogenic response in vitro matching that of the peptide fraction. Other identified peptides point to integrin ligand functionality and more broadly to a diversity of receptor-mediated bioactivity induced by the peptidomic response to nanoparticle exposure. Conclusion The present study demonstrates that pulmonary-sequestered nanoparticles, such as multi-walled carbon nanotubes, acutely upregulate a diverse profile of matrix proteases, and induce a complex peptidomic response across lung and blood compartments. The serum peptide fraction, having cell-surface receptor ligand properties, conveys peripheral bioactivity in promoting endothelial cell inflammation, vasodilatory dysfunction and inhibiting angiogenesis. Results here establish peptide fragments as indirect, non-cytokine mediators and putative biomarkers of systemic health outcomes from nanoparticle exposure.
Collapse
Affiliation(s)
- Ekaterina Mostovenko
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Box 980709, Richmond, VA, 23298-0709, USA
| | - Tamara Young
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Pretal P Muldoon
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Box 980709, Richmond, VA, 23298-0709, USA
| | - Lindsey Bishop
- Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA
| | - Christopher G Canal
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Box 980709, Richmond, VA, 23298-0709, USA
| | - Aleksandar Vucetic
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Box 980709, Richmond, VA, 23298-0709, USA
| | - Patti C Zeidler-Erdely
- Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA
| | - Aaron Erdely
- Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA
| | - Matthew J Campen
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Andrew K Ottens
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Box 980709, Richmond, VA, 23298-0709, USA.
| |
Collapse
|
30
|
Schulte PA, Leso V, Niang M, Iavicoli I. Current state of knowledge on the health effects of engineered nanomaterials in workers: a systematic review of human studies and epidemiological investigations. Scand J Work Environ Health 2019; 45:217-238. [PMID: 30653633 PMCID: PMC6494687 DOI: 10.5271/sjweh.3800] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Objectives The widespread application of nano-enabled products and the increasing likelihood for workplace exposures make understanding engineered nanomaterial (ENM) effects in exposed workers a public and occupational health priority. The aim of this study was to report on the current state of knowledge on possible adverse effects induced by ENM in humans to determine the toxicological profile of each type of ENM and potential biomarkers for early detection of such effects in workers. Methods A systematic review of human studies and epidemiological investigations of exposed workers relative to the possible adverse effects for the most widely used ENM was performed through searches of major scientific databases including Web of Science, Scopus, and PubMed. Results Twenty-seven studies were identified. Most of the epidemiological investigations were cross-sectional. The review found limited evidence of adverse effects in workers exposed to the most commonly used ENM. However, some biological alterations are suggestive for possible adverse impacts. The primary targets of some ENM exposures were the respiratory and cardiovascular systems. Changes in biomarker levels compared with controls were also observed; however, limited exposure data and the relatively short period since the first exposure may have influenced the incidence of adverse effects found in epidemiological studies. Conclusions There is a need for longitudinal epidemiologic investigations with clear exposure characterizations for various ENM to discover potential adverse health effects and identify possible indicators of early biological alterations. In this state of uncertainty, precautionary controls for each ENM are warranted while further study of potential health effects continues.
Collapse
Affiliation(s)
- Paul A Schulte
- National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, 1150 Tusculum Avenue, MS C-14, Cincinnati, OH 45226, USA.
| | | | | | | |
Collapse
|
31
|
Abu Gazia M, El-Magd MA. Effect of pristine and functionalized multiwalled carbon nanotubes on rat renal cortex. Acta Histochem 2019; 121:207-217. [PMID: 30591315 DOI: 10.1016/j.acthis.2018.12.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 12/26/2022]
Abstract
The increasing application of carbon nanotubes (CNTs) within environmental, occupational and consumer settings has raised concerns regarding their biosafety and adverse effects on human health. The present study was designed to investigate the possible adverse effect of pristine and functionalized (amylated and polyethelene glycol coated) multi-walled (MW) CNTs on rat kidney with special concern to the histological alterations and the associated oxidative stress, apoptosis and inflammation. Healthy male albino rats (n = 40) were randomly divided into 4 groups: group I (control), group II (pristine MWCNTs), group III (amylated MWCNTs) and group IV [polyethelene glycol (PEG)-coated MWCNTs]. Animals of groups II, III and IV received a single dose of 1 mg/kg body weight of MWCNTs via intra-tracheal (IT) instillation at the beginning of the experiment and all rats were sacrificed after 30 days. Rats in groups II and III showed, nearly similar, renal tissue damage (evidenced by thin collapsed glomeruli, packed mesangial and endothelial cells as well as edematous hemorrhagic glomeruli with apoptotic changes) and functional disruptions (indicated by high serum levels of urea and creatinine) probably through induction of oxidative stress [revealed by high level of the lipid peroxidation marker malondialdehyde (MDA) and lower levels of the antioxidant enzymes superoxide dismutase (SOD) and glutathione peroxidase (GPx)], apoptosis (indicated by high caspase 3 activity), and inflammation (evidenced by high level of IL1β). However, PEG-coated MWCNTs-treated group (group IV) showed nearly normal renal structure and function. It could be concluded that pristine and functionalized amylated MWCNTs have nephrotoxic effect, while PEG-coated MWCNTs had lowest, or none, toxic effects making them safer for therapy and diagnosis of a variety of diseases.
Collapse
|
32
|
Kan H, Pan D, Castranova V. Engineered nanoparticle exposure and cardiovascular effects: the role of a neuronal-regulated pathway. Inhal Toxicol 2019; 30:335-342. [PMID: 30604639 DOI: 10.1080/08958378.2018.1535634] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Human and animal studies have confirmed that inhalation of particles from ambient air or occupational settings not only causes pathophysiological changes in the respiratory system, but causes cardiovascular effects as well. At an equal mass lung burden, nanoparticles are more potent in causing systemic microvascular dysfunction than fine particles of similar composition. Thus, accumulated evidence from animal studies has led to heightened concerns about the potential short- and long-term deleterious effects of inhalation of engineered nanoparticles on the cardiovascular system. This review highlights the new observations from animal studies, which document the adverse effects of pulmonary exposure to engineered nanoparticles on the cardiovascular system and elucidate the potential mechanisms involved in regulation of cardiovascular function, in particular, how the neuronal system plays a role and reacts to pulmonary nanoparticle exposure based on both in vivo and in vitro studies. In addition, this review also discusses the possible influence of altered autonomic nervous activity on preexisting cardiovascular conditions. Whether engineered nanoparticle exposure serves as a risk factor in the development of cardiovascular diseases warrants further investigation.
Collapse
Affiliation(s)
- H Kan
- a Health Effects Laboratory Division , National Institute for Occupational Safety and Health , Morgantown , WV , USA.,b Department of Pharmaceutical Sciences , West Virginia University , Morgantown , WV , USA
| | - D Pan
- a Health Effects Laboratory Division , National Institute for Occupational Safety and Health , Morgantown , WV , USA
| | - V Castranova
- b Department of Pharmaceutical Sciences , West Virginia University , Morgantown , WV , USA
| |
Collapse
|
33
|
Abstract
Undoubtedly, plastics have changed human existence. These pervasive products are used in nearly every field to include technological, biomedical, and domestic applications. Post-consumer plastic waste disposal leading to plastic pollution in landfills, waterways, and oceans represents a worldwide environmental challenge. Accumulation and continued material fragmentation from micro- to nanoplastics has identified concerns pertaining to environmental and human exposures and toxicity. While many studies have focused on particle fate and identification, the toxicological considerations must focus on the biological relevance of particle deposition within a particular organism, compartment, organ, and tissue. Further, concerns exist regarding the physical and chemical properties of the plastic particles during their production and/or degradation. In this mini-review we will discuss (1) particle characterization and assessment, (2) environmental concerns, and (3) human toxicity.
Collapse
Affiliation(s)
- PA Stapleton
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, 160 Frelinghuysen Rd., Piscataway, NJ 08854, USA
- Environmental and Occupational Health Sciences Institute, 170 Frelinghuysen Rd., Piscataway, NJ 08854, USA
- Correspondence:; Tel: +8484450142
| |
Collapse
|
34
|
Zare-Zardini H, Taheri-Kafrani A, Ordooei M, Amiri A, Karimi-Zarchi M. Evaluation of toxicity of functionalized graphene oxide with ginsenoside Rh2, lysine and arginine on blood cancer cells (K562), red blood cells, blood coagulation and cardiovascular tissue: In vitro and in vivo studies. J Taiwan Inst Chem Eng 2018. [DOI: 10.1016/j.jtice.2018.08.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
35
|
Abukabda AB, McBride CR, Batchelor TP, Goldsmith WT, Bowdridge EC, Garner KL, Friend S, Nurkiewicz TR. Group II innate lymphoid cells and microvascular dysfunction from pulmonary titanium dioxide nanoparticle exposure. Part Fibre Toxicol 2018; 15:43. [PMID: 30413212 PMCID: PMC6230229 DOI: 10.1186/s12989-018-0280-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/24/2018] [Indexed: 01/16/2023] Open
Abstract
Background The cardiovascular effects of pulmonary exposure to engineered nanomaterials (ENM) are poorly understood, and the reproductive consequences are even less understood. Inflammation remains the most frequently explored mechanism of ENM toxicity. However, the key mediators and steps between lung exposure and uterine health remain to be fully defined. The purpose of this study was to determine the uterine inflammatory and vascular effects of pulmonary exposure to titanium dioxide nanoparticles (nano-TiO2). We hypothesized that pulmonary nano-TiO2 exposure initiates a Th2 inflammatory response mediated by Group II innate lymphoid cells (ILC2), which may be associated with an impairment in uterine microvascular reactivity. Methods Female, virgin, Sprague-Dawley rats (8–12 weeks) were exposed to 100 μg of nano-TiO2 via intratracheal instillation 24 h prior to microvascular assessments. Serial blood samples were obtained at 0, 1, 2 and 4 h post-exposure for multiplex cytokine analysis. ILC2 numbers in the lungs were determined. ILC2s were isolated and phosphorylated nuclear factor kappa-light-chain-enhancer of activated B cells (NF-ĸB) levels were measured. Pressure myography was used to assess vascular reactivity of isolated radial arterioles. Results Pulmonary nano-TiO2 exposure was associated with an increase in IL-1ß, 4, 5 and 13 and TNF- α 4 h post-exposure, indicative of an innate Th2 inflammatory response. ILC2 numbers were significantly increased in lungs from exposed animals (1.66 ± 0.19%) compared to controls (0.19 ± 0.22%). Phosphorylation of the transactivation domain (Ser-468) of NF-κB in isolated ILC2 and IL-33 in lung epithelial cells were significantly increased (126.8 ± 4.3% and 137 ± 11% of controls respectively) by nano-TiO2 exposure. Lastly, radial endothelium-dependent arteriolar reactivity was significantly impaired (27 ± 12%), while endothelium-independent dilation (7 ± 14%) and α-adrenergic sensitivity (8 ± 2%) were not altered compared to control levels. Treatment with an anti- IL-33 antibody (1 mg/kg) 30 min prior to nano-TiO2 exposure resulted in a significant improvement in endothelium-dependent dilation and a decreased level of IL-33 in both plasma and bronchoalveolar lavage fluid. Conclusions These results provide evidence that the uterine microvascular dysfunction that follows pulmonary ENM exposure may be initiated via activation of lung-resident ILC2 and subsequent systemic Th2-dependent inflammation. Electronic supplementary material The online version of this article (10.1186/s12989-018-0280-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alaeddin Bashir Abukabda
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Carroll Rolland McBride
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Thomas Paul Batchelor
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - William Travis Goldsmith
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Elizabeth Compton Bowdridge
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Krista Lee Garner
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Sherri Friend
- National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Timothy Robert Nurkiewicz
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA. .,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA. .,National Institute for Occupational Safety and Health, Morgantown, WV, USA.
| |
Collapse
|
36
|
Mandler WK, Nurkiewicz TR, Porter DW, Kelley EE, Olfert IM. Microvascular Dysfunction Following Multiwalled Carbon Nanotube Exposure Is Mediated by Thrombospondin-1 Receptor CD47. Toxicol Sci 2018; 165:90-99. [PMID: 29788500 PMCID: PMC6111784 DOI: 10.1093/toxsci/kfy120] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pulmonary exposure to multiwalled carbon nanotubes (MWCNTs) disrupts peripheral microvascular function. Thrombospondin-1 (TSP-1) is highly expressed during lung injury and has been shown to alter microvascular reactivity. It is unclear exactly how TSP-1 exerts effects on vascular function, but we hypothesized that the TSP-1 receptor CD47 may mediate changes in vasodilation. Wildtype (WT) or CD47 knockout (CD47 KO) C57B6/J-background animals were exposed to 50 µg of MWCNT or saline control via pharyngeal aspiration. Twenty-four hours postexposure, intravital microscopy was performed to assess arteriolar dilation and venular leukocyte adhesion and rolling. To assess tissue redox status, electron paramagnetic resonance and NOx measurements were performed, while inflammatory biomarkers were measured via multiplex assay.Vasodilation was impaired in the WT + MWCNT group compared with control (57 ± 9 vs 90 ± 2% relaxation), while CD47 KO animals showed no impairment (108 ± 8% relaxation). Venular leukocyte adhesion and rolling increased by >2-fold, while the CD47 KO group showed no change. Application of the antioxidant apocynin rescued normal leukocyte activity in the WT + MWCNT group. Lung and plasma NOx were reduced in the WT + MWCNT group by 47% and 32%, respectively, while the CD47 KO groups were unchanged from control. Some inflammatory cytokines were increased in the CD47 + MWCNT group only. In conclusion, TSP-1 is an important ligand mediating MWCNT-induced microvascular dysfunction, and CD47 is a component of this dysregulation. CD47 activation likely disrupts nitric oxide (•NO) signaling and promotes leukocyte-endothelial interactions. Impaired •NO production, signaling, and bioavailability is linked to a variety of cardiovascular diseases in which TSP-1/CD47 may play an important role.
Collapse
Affiliation(s)
- William Kyle Mandler
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV 26506
- Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV 26506
| | - Timothy R Nurkiewicz
- Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV 26506
- Department of Physiology, Pharmacology and Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
- West Virginia Clinical and Translational Science Institute, Robert C. Byrd Health Sciences Center, Morgantown, WV 26506
| | - Dale W Porter
- Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV 26506
- National Institute for Occupational Safety and Health, Morgantown, WV 26505
| | - Eric E Kelley
- Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV 26506
- Department of Physiology, Pharmacology and Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
- West Virginia Clinical and Translational Science Institute, Robert C. Byrd Health Sciences Center, Morgantown, WV 26506
| | - Ivan Mark Olfert
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV 26506
- Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV 26506
- Department of Physiology, Pharmacology and Neuroscience, West Virginia University School of Medicine, Morgantown, WV 26506
- West Virginia Clinical and Translational Science Institute, Robert C. Byrd Health Sciences Center, Morgantown, WV 26506
| |
Collapse
|
37
|
Schulte PA, Kuempel ED, Drew NM. Characterizing risk assessments for the development of occupational exposure limits for engineered nanomaterials. Regul Toxicol Pharmacol 2018; 95:207-219. [PMID: 29574195 PMCID: PMC6075708 DOI: 10.1016/j.yrtph.2018.03.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/05/2018] [Accepted: 03/20/2018] [Indexed: 12/16/2022]
Abstract
The commercialization of engineered nanomaterials (ENMs) began in the early 2000's. Since then the number of commercial products and the number of workers potentially exposed to ENMs is growing, as is the need to evaluate and manage the potential health risks. Occupational exposure limits (OELs) have been developed for some of the first generation of ENMs. These OELs have been based on risk assessments that progressed from qualitative to quantitative as nanotoxicology data became available. In this paper, that progression is characterized. It traces OEL development through the qualitative approach of general groups of ENMs based primarily on read-across with other materials to quantitative risk assessments for nanoscale particles including titanium dioxide, carbon nanotubes and nanofibers, silver nanoparticles, and cellulose nanocrystals. These represent prototypic approaches to risk assessment and OEL development for ENMs. Such substance-by-substance efforts are not practical given the insufficient data for many ENMs that are currently being used or potentially entering commerce. Consequently, categorical approaches are emerging to group and rank ENMs by hazard and potential health risk. The strengths and limitations of these approaches are described, and future derivations and research needs are discussed. Critical needs in moving forward with understanding the health effects of the numerous EMNs include more standardized and accessible quantitative data on the toxicity and physicochemical properties of ENMs.
Collapse
Affiliation(s)
- P A Schulte
- National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, United States.
| | - E D Kuempel
- National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, United States
| | - N M Drew
- National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, United States
| |
Collapse
|
38
|
Stapleton PA, McBride CR, Yi J, Abukabda AB, Nurkiewicz TR. Estrous cycle-dependent modulation of in vivo microvascular dysfunction after nanomaterial inhalation. Reprod Toxicol 2018; 78:20-28. [PMID: 29545171 PMCID: PMC6034709 DOI: 10.1016/j.reprotox.2018.03.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 02/08/2018] [Accepted: 03/05/2018] [Indexed: 12/11/2022]
Abstract
Preconceptive health encompasses male and female reproductive capability. In females, this takes into account each of the stages of the estrous cycle. Microvascular reactivity varies throughout the estrous cycle in response to hormonal changes and in preparation for pregnancy. Microvascular alterations in response to engineered nanomaterial (ENM) exposure have been described within 24-h of inhalation; however, the impact upon the uterine vasculature at differing estrous stages and at late-stage pregnancy is unclear. Female Sprague Dawley (SD) rats (virgin and late stage pregnancy [GD 19]) were exposed to nano-TiO aerosols (173.2 ± 6.4 nm, 10.2 ± 0.46 mg/m3, 5 h) 24-h prior to experimentation leading to a single calculated deposition of 42.2 ± 1.9 µg nano- TiO2 (exposed) or 0µg (control). Animals were anesthetized, estrous status verified, and prepared for in situ assessment of leukocyte trafficking and vascular function by means of intravital microscopy, Uterine basal arteriolar reactivity was stimulated using iontophoretically applied chemicals: acetylcholine (ACh, 0.025 M; 20, 40, 100, 200 nA), sodium nitroprusside (SNP, 0.05 M; 20, 40, 100 nA), phenylephrine (PE, 0.05 M; 20, 40, 100 nA). Finally, adenosine (ADO, 10−4 M) was superfused over the tissue to identify maximum diameter. In situ vessel reactivity after exposure was significantly blunted based on estrous stage, but not at late-stage pregnancy. Local uterine venular leukocyte trafficking and systemic inflammatory markers were also significantly affected during preparatory (proestrus), fertile (estrus), and infertile (diestrus) periods after ENM inhalation. Overall, these deficits in reactivity and increased inflammatory activity may impair female fertility after ENM exposure.
Collapse
Affiliation(s)
- P A Stapleton
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA; Environmental and Occupational Health Sciences Institute, Piscataway, NJ, USA.
| | - C R McBride
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University, Morgantown, WV, USA; Toxicology Working Group, West Virginia University, Morgantown, WV, USA
| | - J Yi
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University, Morgantown, WV, USA
| | - A B Abukabda
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University, Morgantown, WV, USA; Toxicology Working Group, West Virginia University, Morgantown, WV, USA
| | - T R Nurkiewicz
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University, Morgantown, WV, USA; Toxicology Working Group, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
39
|
Stapleton PA, Hathaway QA, Nichols CE, Abukabda AB, Pinti MV, Shepherd DL, McBride CR, Yi J, Castranova VC, Hollander JM, Nurkiewicz TR. Maternal engineered nanomaterial inhalation during gestation alters the fetal transcriptome. Part Fibre Toxicol 2018; 15:3. [PMID: 29321036 PMCID: PMC5763571 DOI: 10.1186/s12989-017-0239-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 12/21/2017] [Indexed: 01/19/2023] Open
Abstract
Background The integration of engineered nanomaterials (ENM) is well-established and widespread in clinical, commercial, and domestic applications. Cardiovascular dysfunctions have been reported in adult populations after exposure to a variety of ENM. As the diversity of these exposures continues to increase, the fetal ramifications of maternal exposures have yet to be determined. We, and others, have explored the consequences of ENM inhalation during gestation and identified many cardiovascular and metabolic outcomes in the F1 generation. The purpose of these studies was to identify genetic alterations in the F1 generation of Sprague-Dawley rats that result from maternal ENM inhalation during gestation. Pregnant dams were exposed to nano-titanium dioxide (nano-TiO2) aerosols (10 ± 0.5 mg/m3) for 7-8 days (calculated, cumulative lung deposition = 217 ± 1 μg) and on GD (gestational day) 20 fetal hearts were isolated. DNA was extracted and immunoprecipitated with modified chromatin marks histone 3 lysine 4 tri-methylation (H3K4me3) and histone 3 lysine 27 tri-methylation (H3K27me3). Following chromatin immunoprecipitation (ChIP), DNA fragments were sequenced. RNA from fetal hearts was purified and prepared for RNA sequencing and transcriptomic analysis. Ingenuity Pathway Analysis (IPA) was then used to identify pathways most modified by gestational ENM exposure. Results The results of the sequencing experiments provide initial evidence that significant epigenetic and transcriptomic changes occur in the cardiac tissue of maternal nano-TiO2 exposed progeny. The most notable alterations in major biologic systems included immune adaptation and organismal growth. Changes in normal physiology were linked with other tissues, including liver and kidneys. Conclusions These results are the first evidence that maternal ENM inhalation impacts the fetal epigenome. Electronic supplementary material The online version of this article (10.1186/s12989-017-0239-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- P A Stapleton
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA.,Environmental and Occupational Health Sciences Institute, Piscataway, NJ, USA
| | - Q A Hathaway
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA.,Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - C E Nichols
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - A B Abukabda
- Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA.,Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, USA
| | - M V Pinti
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA.,Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - D L Shepherd
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA.,Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - C R McBride
- Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA.,Department of Physiology, Pharmacology, and Neuroscience, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, 1 Medical Center Drive, Morgantown, WV, 26506-9229, USA
| | - J Yi
- Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA.,Department of Physiology, Pharmacology, and Neuroscience, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, 1 Medical Center Drive, Morgantown, WV, 26506-9229, USA
| | - V C Castranova
- Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA.,Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, USA
| | - J M Hollander
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA.,Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - T R Nurkiewicz
- Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, WV, USA. .,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA. .,Department of Physiology, Pharmacology, and Neuroscience, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, 1 Medical Center Drive, Morgantown, WV, 26506-9229, USA.
| |
Collapse
|
40
|
Inhalation exposure to three-dimensional printer emissions stimulates acute hypertension and microvascular dysfunction. Toxicol Appl Pharmacol 2017; 335:1-5. [PMID: 28942003 DOI: 10.1016/j.taap.2017.09.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 09/19/2017] [Indexed: 01/19/2023]
Abstract
Fused deposition modeling (FDM™), or three-dimensional (3D) printing has become routine in industrial, occupational and domestic environments. We have recently reported that 3D printing emissions (3DPE) are complex mixtures, with a large ultrafine particulate matter component. Additionally, we and others have reported that inhalation of xenobiotic particles in this size range is associated with an array of cardiovascular dysfunctions. Sprague-Dawley rats were exposed to 3DPE aerosols via nose-only exposure for ~3h. Twenty-four hours later, intravital microscopy was performed to assess microvascular function in the spinotrapezius muscle. Endothelium-dependent and -independent arteriolar dilation were stimulated by local microiontophoresis of acetylcholine (ACh) and sodium nitroprusside (SNP). At the time of experiments, animals exposed to 3DPE inhalation presented with a mean arterial pressure of 125±4mmHg, and this was significantly higher than that for the sham-control group (94±3mmHg). Consistent with this pressor response in the 3DPE group, was an elevation of ~12% in resting arteriolar tone. Endothelium-dependent arteriolar dilation was significantly impaired after 3DPE inhalation across all iontophoretic ejection currents (0-27±15%, compared to sham-control: 15-120±21%). Endothelium-independent dilation was not affected by 3DPE inhalation. These alterations in peripheral microvascular resistance and reactivity are consistent with elevations in arterial pressure that follow 3DPE inhalation. Future studies must identify the specific toxicants generated by FDM™ that drive this acute pressor response.
Collapse
|
41
|
Abukabda AB, Stapleton PA, McBride CR, Yi J, Nurkiewicz TR. Heterogeneous Vascular Bed Responses to Pulmonary Titanium Dioxide Nanoparticle Exposure. Front Cardiovasc Med 2017; 4:33. [PMID: 28596957 PMCID: PMC5442182 DOI: 10.3389/fcvm.2017.00033] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/01/2017] [Indexed: 01/06/2023] Open
Abstract
A growing body of research links engineered nanomaterial (ENM) exposure to adverse cardiovascular endpoints. The purpose of this study was to evaluate the impact of ENM exposure on vascular reactivity in discrete segments so that we may determine the most sensitive levels of the vasculature where these negative cardiovascular effects are manifest. We hypothesized that acute nano-TiO2 exposure differentially affects reactivity with a more robust impairment in the microcirculation. Sprague-Dawley rats (8–10 weeks) were exposed to nano-TiO2via intratracheal instillation (20, 100, or 200 µg suspended per 250 µL of vehicle) 24 h prior to vascular assessments. A serial assessment across distinct compartments of the vascular tree was then conducted. Wire myography was used to evaluate macrovascular active tension generation specifically in the thoracic aorta, the femoral artery, and third-order mesenteric arterioles. Pressure myography was used to determine vascular reactivity in fourth- and fifth-order mesenteric arterioles. Vessels were treated with phenylephrine, acetylcholine (ACh), and sodium nitroprusside. Nano-TiO2 exposure decreased endothelium-dependent relaxation in the thoracic aorta and femoral arteries assessed via ACh by 53.96 ± 11.6 and 25.08 ± 6.36%, respectively. Relaxation of third-order mesenteric arterioles was impaired by 100 and 20 µg nano-TiO2 exposures with mean reductions of 50.12 ± 8.7 and 68.28 ± 8.7%. Cholinergic reactivity of fourth- and fifth-order mesenteric arterioles was negatively affected by nano-TiO2 with diminished dilations of 82.86 ± 12.6% after exposure to 200 µg nano-TiO2, 42.6 ± 12.6% after 100 µg nano-TiO2, and 49.4 ± 12.6% after 20 µg nano-TiO2. Endothelium-independent relaxation was impaired in the thoracic aorta by 34.05 ± 25% induced by exposure to 200 µg nano-TiO2 and a reduction in response of 49.31 ± 25% caused by 100 µg nano-TiO2. Femoral artery response was reduced by 18 ± 5%, while third-order mesenteric arterioles were negatively affected by 20 µg nano-TiO2 with a mean decrease in response of 38.37 ± 10%. This is the first study to directly compare the differential effect of ENM exposure on discrete anatomical segments of the vascular tree. Pulmonary ENM exposure produced macrovascular and microvascular dysfunction resulting in impaired responses to endothelium-dependent, endothelium-independent, and adrenergic agonists with a more robust dysfunction at the microvascular level. These results provide additional evidence of an endothelium-dependent and endothelium-independent impairment in vascular reactivity.
Collapse
Affiliation(s)
- Alaeddin B Abukabda
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Phoebe A Stapleton
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA
| | - Carroll R McBride
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Jinghai Yi
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Timothy R Nurkiewicz
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
42
|
Poulsen SS, Knudsen KB, Jackson P, Weydahl IEK, Saber AT, Wallin H, Vogel U. Multi-walled carbon nanotube-physicochemical properties predict the systemic acute phase response following pulmonary exposure in mice. PLoS One 2017; 12:e0174167. [PMID: 28380028 PMCID: PMC5381870 DOI: 10.1371/journal.pone.0174167] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/04/2017] [Indexed: 01/08/2023] Open
Abstract
Pulmonary exposure to multi-walled carbon nanotubes (MWCNTs) has been linked to an increased risk of developing cardiovascular disease in addition to the well-documented physicochemical-dependent adverse lung effects. A proposed mechanism is through a strong and sustained pulmonary secretion of acute phase proteins to the blood. We identified physicochemical determinants of MWCNT-induced systemic acute phase response by analyzing effects of pulmonary exposure to 14 commercial, well-characterized MWCNTs in female C57BL/6J mice pulmonary exposed to 0, 6, 18 or 54 μg MWCNT/mouse. Plasma levels of acute phase response proteins serum amyloid A1/2 (SAA1/2) and SAA3 were determined on day 1, 28 or 92. Expression levels of hepatic Saa1 and pulmonary Saa3 mRNA levels were assessed to determine the origin of the acute phase response proteins. Pulmonary Saa3 mRNA expression levels were greater and lasted longer than hepatic Saa1 mRNA expression. Plasma SAA1/2 and SAA3 protein levels were related to time and physicochemical properties using adjusted, multiple regression analyses. SAA3 and SAA1/2 plasma protein levels were increased after exposure to almost all of the MWCNTs on day 1, whereas limited changes were observed on day 28 and 92. SAA1/2 and SAA3 protein levels did not correlate and only SAA3 protein levels correlated with neutrophil influx. The multiple regression analyses revealed a protective effect of MWCNT length on SAA1/2 protein level on day 1, such that a longer length resulted in lowered SAA1/2 plasma levels. Increased SAA3 protein levels were positively related to dose and content of Mn, Mg and Co on day 1, whereas oxidation and diameter of the MWCNTs were protective on day 28 and 92, respectively. The results of this study reveal very differently controlled pulmonary and hepatic acute phase responses after MWCNT exposure. As the responses were influenced by the physicochemical properties of the MWCNTs, this study provides the first step towards designing MWCNT that induce less SAA.
Collapse
Affiliation(s)
- Sarah S. Poulsen
- National Research Centre for the Working Environment, Copenhagen Ø, Denmark
- * E-mail:
| | | | - Petra Jackson
- National Research Centre for the Working Environment, Copenhagen Ø, Denmark
| | | | - Anne T. Saber
- National Research Centre for the Working Environment, Copenhagen Ø, Denmark
| | - Håkan Wallin
- National Research Centre for the Working Environment, Copenhagen Ø, Denmark
- Institute of Public Health, Copenhagen University, Copenhagen K, Denmark
| | - Ulla Vogel
- National Research Centre for the Working Environment, Copenhagen Ø, Denmark
- Department of Micro- and Nanotechnology, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
43
|
Mandler WK, Nurkiewicz TR, Porter DW, Olfert IM. Thrombospondin-1 mediates multi-walled carbon nanotube induced impairment of arteriolar dilation. Nanotoxicology 2017; 11:112-122. [PMID: 28024456 DOI: 10.1080/17435390.2016.1277275] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pulmonary exposure to multi-walled carbon nanotubes (MWCNT) has been shown to disrupt endothelium-dependent arteriolar dilation in the peripheral microcirculation. The molecular mechanisms behind these arteriolar disruptions have yet to be fully elucidated. The secreted matricellular matrix protein thrombospondin-1 (TSP-1) is capable of moderating arteriolar vasodilation by inhibiting soluble guanylate cyclase activity. We hypothesized that TSP-1 may be a link between nanomaterial exposure and observed peripheral microvascular dysfunction. To test this hypothesis, wild-type C57B6J (WT) and TSP-1 knockout (KO) mice were exposed via lung aspiration to 50 μg MWCNT or a Sham dispersion medium control. Following exposure (24 h), arteriolar characteristics and reactivity were measured in the gluteus maximus muscle using intravital microscopy (IVM) coupled with microiontophoretic delivery of acetylcholine (ACh) or sodium nitroprusside (SNP). In WT mice exposed to MWCNT, skeletal muscle TSP-1 protein increased > fivefold compared to Sham exposed, and exhibited a 39% and 47% decrease in endothelium-dependent and -independent vasodilation, respectively. In contrast, TSP-1 protein was not increased following MWCNT exposure in KO mice and exhibited no loss in dilatory capacity. Microvascular leukocyte-endothelium interactions were measured by assessing leukocyte adhesion and rolling activity in third order venules. The WT + MWCNT group demonstrated 223% higher leukocyte rolling compared to the WT + Sham controls. TSP-1 KO animals exposed to MWCNT showed no differences from the WT + Sham control. These data provide evidence that TSP-1 is likely a central mediator of the systemic microvascular dysfunction that follows pulmonary MWCNT exposure.
Collapse
Affiliation(s)
- W Kyle Mandler
- a Division of Exercise Physiology , West Virginia University School of Medicine , Morgantown , WV , USA
| | - Timothy R Nurkiewicz
- b Department of Physiology and Pharmacology , West Virginia University School of Medicine , Morgantown , WV , USA.,c Center for Cardiovascular & Respiratory Sciences , West Virginia University, Robert C. Byrd Health Sciences Center , Morgantown , WV , USA
| | - Dale W Porter
- d National Institute for Occupational Safety and Health , Morgantown , WV , USA
| | - I Mark Olfert
- a Division of Exercise Physiology , West Virginia University School of Medicine , Morgantown , WV , USA.,c Center for Cardiovascular & Respiratory Sciences , West Virginia University, Robert C. Byrd Health Sciences Center , Morgantown , WV , USA
| |
Collapse
|
44
|
Hathaway QA, Nichols CE, Shepherd DL, Stapleton PA, McLaughlin SL, Stricker JC, Rellick SL, Pinti MV, Abukabda AB, McBride CR, Yi J, Stine SM, Nurkiewicz TR, Hollander JM. Maternal-engineered nanomaterial exposure disrupts progeny cardiac function and bioenergetics. Am J Physiol Heart Circ Physiol 2016; 312:H446-H458. [PMID: 28011589 PMCID: PMC5402018 DOI: 10.1152/ajpheart.00634.2016] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/07/2016] [Accepted: 12/07/2016] [Indexed: 01/25/2023]
Abstract
Nanomaterial production is expanding as new industrial and consumer applications are introduced. Nevertheless, the impacts of exposure to these compounds are not fully realized. The present study was designed to determine whether gestational nano-sized titanium dioxide exposure impacts cardiac and metabolic function of developing progeny. Pregnant Sprague-Dawley rats were exposed to nano-aerosols (~10 mg/m3, 130- to 150-nm count median aerodynamic diameter) for 7-8 nonconsecutive days, beginning at gestational day 5-6 Physiological and bioenergetic effects on heart function and cardiomyocytes across three time points, fetal (gestational day 20), neonatal (4-10 days), and young adult (6-12 wk), were evaluated. Functional analysis utilizing echocardiography, speckle-tracking based strain, and cardiomyocyte contractility, coupled with mitochondrial energetics, revealed effects of nano-exposure. Maternal exposed progeny demonstrated a decrease in E- and A-wave velocities, with a 15% higher E-to-A ratio than controls. Myocytes isolated from exposed animals exhibited ~30% decrease in total contractility, departure velocity, and area of contraction. Bioenergetic analysis revealed a significant increase in proton leak across all ages, accompanied by decreases in metabolic function, including basal respiration, maximal respiration, and spare capacity. Finally, electron transport chain complex I and IV activities were negatively impacted in the exposed group, which may be linked to a metabolic shift. Molecular data suggest that an increase in fatty acid metabolism, uncoupling, and cellular stress proteins may be associated with functional deficits of the heart. In conclusion, gestational nano-exposure significantly impairs the functional capabilities of the heart through cardiomyocyte impairment, which is associated with mitochondrial dysfunction.NEW & NOTEWORTHY Cardiac function is evaluated, for the first time, in progeny following maternal nanomaterial inhalation. The findings indicate that exposure to nano-sized titanium dioxide (nano-TiO2) during gestation negatively impacts cardiac function and mitochondrial respiration and bioenergetics. We conclude that maternal nano-TiO2 inhalation contributes to adverse cardiovascular health effects, lasting into adulthood.
Collapse
Affiliation(s)
- Quincy A Hathaway
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia.,Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Cody E Nichols
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Danielle L Shepherd
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia.,Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Phoebe A Stapleton
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Sarah L McLaughlin
- Department of Cancer Cell Biology, West Virginia University School of Medicine; Morgantown, West Virginia; and
| | - Janelle C Stricker
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Stephanie L Rellick
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Mark V Pinti
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Alaeddin B Abukabda
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Carroll R McBride
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Jinghai Yi
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Seth M Stine
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia.,Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Timothy R Nurkiewicz
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia.,Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, West Virginia
| | - John M Hollander
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, West Virginia; .,Mitochondria, Metabolism & Bioenergetics Working Group, West Virginia University School of Medicine, Morgantown, West Virginia
| |
Collapse
|
45
|
Bostan HB, Rezaee R, Valokala MG, Tsarouhas K, Golokhvast K, Tsatsakis AM, Karimi G. Cardiotoxicity of nano-particles. Life Sci 2016; 165:91-99. [PMID: 27686832 DOI: 10.1016/j.lfs.2016.09.017] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 09/14/2016] [Accepted: 09/23/2016] [Indexed: 01/08/2023]
Abstract
Nano-particles (NPs) are used in industrial and biomedical fields such as cosmetics, food additives and biosensors. Beside their favorable properties, nanoparticles are responsible for toxic effects. Local adverse effects and/or systemic toxicity are described with nanoparticle delivery to target organs of the human body. Animal studies provide evidence for the aforementioned toxicity. Cardiac function is a specific target of nanoparticles. Thus, reviewing the current bibliography on cardiotoxicity of nanoparticles and specifically of titanium, zinc, silver, carbon, silica and iron oxide nano-materials is the aim of this study.
Collapse
Affiliation(s)
- Hasan Badie Bostan
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ramin Rezaee
- Department of Physiology and Pharmacology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Scientific Educational Center of Nanotechnology, Far Eastern Federal University, 10 Pushkinskaya Street, Vladivostok 690950, Russia
| | - Mahmoud Gorji Valokala
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Kirill Golokhvast
- Scientific Educational Center of Nanotechnology, Far Eastern Federal University, 10 Pushkinskaya Street, Vladivostok 690950, Russia
| | - Aristidis M Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, Heraklion 71003, Greece.
| | - Gholamreza Karimi
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
46
|
Secondo LE, Liu NJ, Lewinski NA. Methodological considerations when conductingin vitro, air–liquid interface exposures to engineered nanoparticle aerosols. Crit Rev Toxicol 2016; 47:225-262. [DOI: 10.1080/10408444.2016.1223015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Lynn E. Secondo
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Nathan J. Liu
- Institute for Work and Health (IST), University of Lausanne and Geneva, Epalinges-Lausanne, Switzerland
- Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Nastassja A. Lewinski
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, VA, USA
- Institute for Work and Health (IST), University of Lausanne and Geneva, Epalinges-Lausanne, Switzerland
| |
Collapse
|
47
|
Naresh NK, Butcher JT, Lye RJ, Chen X, Isakson BE, Gan LM, Kramer CM, Annex BH, Epstein FH. Cardiovascular magnetic resonance detects the progression of impaired myocardial perfusion reserve and increased left-ventricular mass in mice fed a high-fat diet. J Cardiovasc Magn Reson 2016; 18:53. [PMID: 27609091 PMCID: PMC5016874 DOI: 10.1186/s12968-016-0273-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 08/11/2016] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Impaired myocardial perfusion reserve (MPR) is prevalent in obesity and diabetes, even in the absence of obstructive coronary artery disease (CAD), and is prognostic of adverse events. We sought to establish the time course of reduced MPR and to investigate associated vascular and tissue properties in mice fed a high-fat diet (HFD), as they are an emerging model of human obesity, diabetes, and reduced MPR without obstructive CAD. METHODS C57Bl/6 mice fed a HFD or a low-fat diet (control) were imaged at 6, 12, 18 and 24 weeks post-diet. The cardiovascular magnetic resonance (CMR) protocol included multi-slice cine imaging to assess ejection fraction (EF), left-ventricular (LV) mass, LV wall thickness (LVWT), and LV volumes, and first-pass perfusion CMR to quantify MPR. Coronary vascular reactivity, aortic atherosclerosis, myocardial capillary density and tissue fibrosis were also assessed. RESULTS Body weight was increased in HFD mice at 6-24 weeks post-diet (p < 0.05 vs. control). MPR in HFD mice was reduced and LV mass and LVWT were increased in HFD mice at 18 and 24 weeks post-diet (p < 0.05 vs. control). Coronary arteriolar vascular reactivity to adenosine and acetylcholine were reduced in HFD mice (p < 0.05 vs. control). There were no significant differences in cardiac volumes, EF, or capillary density measurements between the two groups. Histology showed interstitial fibrosis in HFD and no aortic atherosclerosis in either group. CONCLUSIONS C57Bl/6 mice fed a HFD for 18-24 weeks have progressively increased LV mass and impaired MPR with fibrosis, normal capillary density and no aortic plaque. These results establish C57Bl/6 mice fed a HFD for 18-24 weeks as a model of impaired MPR without obstructive CAD due to obesity and diabetes.
Collapse
Affiliation(s)
- Nivedita K. Naresh
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA USA
| | - Joshua T. Butcher
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA USA
| | - Robert J. Lye
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA USA
| | - Xiao Chen
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA USA
| | - Brant E. Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA USA
| | - Li-Ming Gan
- Department of Molecular and Clinical Medicine, AstraZeneca R&D, Mölndal, Sweden
- Institute of Medicine, Sahlgrenska Academy, CVMD Early Clinical Development, AstraZeneca R&D, Mölndal, Sweden
| | - Christopher M. Kramer
- Cardiovascular Medicine, University of Virginia, Charlottesville, VA USA
- Department of Radiology, University of Virginia, Charlottesville, VA USA
| | - Brian H. Annex
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA USA
- Cardiovascular Medicine, University of Virginia, Charlottesville, VA USA
| | - Frederick H. Epstein
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA USA
- Department of Radiology, University of Virginia, Charlottesville, VA USA
| |
Collapse
|
48
|
Ruenraroengsak P, Chen S, Hu S, Melbourne J, Sweeney S, Thorley AJ, Skepper JN, Shaffer MSP, Tetley TD, Porter AE. Translocation of Functionalized Multi-Walled Carbon Nanotubes across Human Pulmonary Alveolar Epithelium: Dominant Role of Epithelial Type 1 Cells. ACS NANO 2016; 10:5070-85. [PMID: 27035850 PMCID: PMC6682507 DOI: 10.1021/acsnano.5b08218] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Uptake and translocation of short functionalized multi-walled carbon nanotubes (short-fMWCNTs) through the pulmonary respiratory epithelial barrier depend on physicochemical property and cell type. Two monoculture models, immortalized human alveolar epithelial type 1 (TT1) cells and primary human alveolar epithelial type 2 cells (AT2), which constitute the alveolar epithelial barrier, were employed to investigate the uptake and transport of 300 and 700 nm in length, poly(4-vinylpyridine)-functionalized, multi-walled carbon nanotubes (p(4VP)-MWCNTs) using quantitative imaging and spectroscopy techniques. The p(4VP)-MWCNT exhibited no toxicity on TT1 and AT2 cells, but significantly decreased barrier integrity (*p < 0.01). Uptake of p(4VP)-MWCNTs was observed in 70% of TT1 cells, correlating with compromised barrier integrity and basolateral p(4VP)-MWCNT translocation. There was a small but significantly greater uptake of 300 nm p(4VP)-MWCNTs than 700 nm p(4VP)-MWCNTs by TT1 cells. Up to 3% of both the 300 and 700 nm p(4VP)-MWCNTs reach the basal chamber; this relatively low amount arose because the supporting transwell membrane minimized the amount of p(4VP)-MWCNT translocating to the basal chamber, seen trapped between the basolateral cell membrane and the membrane. Only 8% of AT2 cells internalized p(4VP)-MWCNT, accounting for 17% of applied p(4VP)-MWCNT), with transient effects on barrier function, which initially fell then returned to normal; there was no MWCNT basolateral translocation. The transport rate was MWCNT length modulated. The comparatively lower p(4VP)-MWCNT uptake by AT2 cells is proposed to reflect a primary barrier effect of type 2 cell secretions and the functional differences between the type 1 and type 2 alveolar epithelial cells.
Collapse
Affiliation(s)
- Pakatip Ruenraroengsak
- Department of Materials and London Centre for Nanotechnology, Imperial College London, Exhibition Road, London, UK, SW7 2AZ
- Lung Cell Biology, Airways Disease, National Heart and Lung Institute, Imperial College London, Dovehouse Street, London, UK, SW3 6LY
- Correspondence should be addressed to: Dr Pakatip Ruenraroengsak, Department of Materials and London Centre for Nanotechnology, Imperial College London, London SW7 2AZ, UK, ; Prof. Teresa D. Tetley, Department of Lung Cell Biology, National Heart and Lung Institute, Guy Scadding Building, Dovehouse Street, London SW3 6LY, UK Phone: +44-207-5942984, ; Dr Alexandra E. Porter, Department of Materials and London Centre for Nanotechnology, Imperial College London, London SW7 2AZ, UK, Phone: +44-207-594691,
| | - Shu Chen
- Department of Materials and London Centre for Nanotechnology, Imperial College London, Exhibition Road, London, UK, SW7 2AZ
| | - Sheng Hu
- Department of Chemistry and London Centre for Nanotechnology, Imperial College London, Exhibition Road, London, UK
| | - Jodie Melbourne
- Department of Materials and London Centre for Nanotechnology, Imperial College London, Exhibition Road, London, UK, SW7 2AZ
| | - Sinbad Sweeney
- Lung Cell Biology, Airways Disease, National Heart and Lung Institute, Imperial College London, Dovehouse Street, London, UK, SW3 6LY
| | - Andrew J. Thorley
- Lung Cell Biology, Airways Disease, National Heart and Lung Institute, Imperial College London, Dovehouse Street, London, UK, SW3 6LY
| | - Jeremy N. Skepper
- Cambridge Advanced Imaging Centre, Department of Physiology, Development and Neuroscience, University of Cambridge, UK, CB2 3DY
| | - Milo S. P. Shaffer
- Department of Chemistry and London Centre for Nanotechnology, Imperial College London, Exhibition Road, London, UK
| | - Teresa D. Tetley
- Lung Cell Biology, Airways Disease, National Heart and Lung Institute, Imperial College London, Dovehouse Street, London, UK, SW3 6LY
- Correspondence should be addressed to: Dr Pakatip Ruenraroengsak, Department of Materials and London Centre for Nanotechnology, Imperial College London, London SW7 2AZ, UK, ; Prof. Teresa D. Tetley, Department of Lung Cell Biology, National Heart and Lung Institute, Guy Scadding Building, Dovehouse Street, London SW3 6LY, UK Phone: +44-207-5942984, ; Dr Alexandra E. Porter, Department of Materials and London Centre for Nanotechnology, Imperial College London, London SW7 2AZ, UK, Phone: +44-207-594691,
| | - Alexandra E. Porter
- Department of Materials and London Centre for Nanotechnology, Imperial College London, Exhibition Road, London, UK, SW7 2AZ
- Correspondence should be addressed to: Dr Pakatip Ruenraroengsak, Department of Materials and London Centre for Nanotechnology, Imperial College London, London SW7 2AZ, UK, ; Prof. Teresa D. Tetley, Department of Lung Cell Biology, National Heart and Lung Institute, Guy Scadding Building, Dovehouse Street, London SW3 6LY, UK Phone: +44-207-5942984, ; Dr Alexandra E. Porter, Department of Materials and London Centre for Nanotechnology, Imperial College London, London SW7 2AZ, UK, Phone: +44-207-594691,
| |
Collapse
|
49
|
Møller P, Christophersen DV, Jacobsen NR, Skovmand A, Gouveia ACD, Andersen MHG, Kermanizadeh A, Jensen DM, Danielsen PH, Roursgaard M, Jantzen K, Loft S. Atherosclerosis and vasomotor dysfunction in arteries of animals after exposure to combustion-derived particulate matter or nanomaterials. Crit Rev Toxicol 2016; 46:437-76. [DOI: 10.3109/10408444.2016.1149451] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
50
|
Thompson LC, Holland NA, Snyder RJ, Luo B, Becak DP, Odom JT, Harrison BS, Brown JM, Gowdy KM, Wingard CJ. Pulmonary instillation of MWCNT increases lung permeability, decreases gp130 expression in the lungs, and initiates cardiovascular IL-6 transsignaling. Am J Physiol Lung Cell Mol Physiol 2015; 310:L142-54. [PMID: 26589480 DOI: 10.1152/ajplung.00384.2014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 11/06/2015] [Indexed: 12/24/2022] Open
Abstract
Pulmonary instillation of multiwalled carbon nanotubes (MWCNT) has the potential to promote cardiovascular derangements, but the mechanisms responsible are currently unclear. We hypothesized that exposure to MWCNT would result in increased epithelial barrier permeability by 24 h postexposure and initiate a signaling process involving IL-6/gp130 transsignaling in peripheral vascular tissue. To test this hypothesis we assessed the impact of 1 and 10 μg/cm(2) MWCNT on transepithelial electrical resistance (TEER) and expression of barrier proteins and cell activation in vitro using normal human bronchial epithelial primary cells. Parallel studies using male Sprague-Dawley rats instilled with 100 μg MWCNT measured bronchoalveolar lavage (BAL) differential cell counts, BAL fluid total protein, and lung water-to-tissue weight ratios 24 h postexposure and quantified serum concentrations of IL-6, soluble IL-6r, and soluble gp130. Aortic sections were examined immunohistochemically for gp130 expression, and gp130 mRNA/protein expression was evaluated in rat lung, heart, and aortic tissue homogenates. Our in vitro findings indicate that 10 μg/cm(2) MWCNT decreased the development of TEER and zonula occludens-1 expression relative to the vehicle. In rats MWCNT instillation increased BAL protein, lung water, and induced pulmonary eosinophilia. Serum concentrations of soluble gp130 decreased, aortic endothelial expression of gp130 increased, and expression of gp130 in the lung was downregulated in the MWCNT-exposed group. We propose that pulmonary exposure to MWCNT can manifest as a reduced epithelial barrier and activator of vascular gp130-associated transsignaling that may promote susceptibility to cardiovascular derangements.
Collapse
Affiliation(s)
- Leslie C Thompson
- Department of Physiology, Brody School of Medicine at East Carolina University, Greenville, North Carolina
| | - Nathan A Holland
- Department of Physiology, Brody School of Medicine at East Carolina University, Greenville, North Carolina
| | - Ryan J Snyder
- NanoHealth Program, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina; and
| | - Bin Luo
- Department of Pharmacology & Toxicology, Brody School of Medicine at East Carolina University, Greenville, North Carolina
| | - Daniel P Becak
- Department of Physiology, Brody School of Medicine at East Carolina University, Greenville, North Carolina
| | - Jillian T Odom
- Department of Physiology, Brody School of Medicine at East Carolina University, Greenville, North Carolina
| | - Benjamin S Harrison
- Wake Forest University Institute of Regenerative Medicine, Winston-Salem, North Carolina
| | - Jared M Brown
- Department of Pharmacology & Toxicology, Brody School of Medicine at East Carolina University, Greenville, North Carolina
| | - Kymberly M Gowdy
- Department of Pharmacology & Toxicology, Brody School of Medicine at East Carolina University, Greenville, North Carolina
| | - Christopher J Wingard
- Department of Physiology, Brody School of Medicine at East Carolina University, Greenville, North Carolina;
| |
Collapse
|