1
|
DeVallance E, Bowdridge E, Garner K, Griffith J, Seman M, Batchelor T, Velayutham M, Goldsmith WT, Hussain S, Kelley EE, Nurkiewicz TR. The alarmin, interleukin-33, increases vascular tone via extracellular signal regulated kinase-mediated Ca 2+ sensitization and endothelial nitric oxide synthase inhibition. J Physiol 2024; 602:6087-6107. [PMID: 39540837 DOI: 10.1113/jp286990] [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: 05/28/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Alarmins are classified by their release from damaged or ruptured cells. Many alarmins have been found to increase vascular tone and oppose endothelium-dependent dilatation (EDD). Interleukin (IL)-33 plays a prominent role in lung injury and can be released during vascular injury and in chronic studies found to be cardioprotective. Our recent work has implicated IL-33 in acute vascular dysfunction following inhalation of engineered nanomaterials (ENM). However, the mechanisms linking IL-33 to vascular tone have not been interrogated. We therefore aimed to determine whether IL-33 directly influenced microvascular tone and endothelial function. Isolated feed arteries and in vivo arterioles from male and female Sprague-Dawley rats were used to determine direct vascular actions of IL-33. Mesenteric feed arteries and arterioles demonstrated reduced intraluminal diameters when treated with increasing concentrations of recombinant IL-33. IL-33 activated extracellular signal regulated kinase (ERK)1/2 of rat aortic smooth muscle cells but not phosphorylation of myosin light chain kinase. This suggested IL-33 may sensitize arterioles to Ca2+-mediated responses. Indeed, IL-33 augmented the myogenic- and phenylephrine-induced vasoconstriction. Additionally, incubation of arterioles with 1 ng IL-33 attenuated ACh-mediated EDD. Mechanistically, in human aortic endothelial cells, we demonstrate that IL-33-mediated ERK1/2 activation leads to inhibitory phosphorylation of serine 602 on endothelial nitric oxide synthase. Finally, we demonstrate that IL-33-ERK1/2 contributes to vascular tone following two known inducers of IL-33; ENM inhalation and the rupture endothelial cells. The present study provides novel evidence that IL-33 increases vascular tone via canonical ERK1/2 activation in microvascular smooth muscle and endothelium. Altogether, it is suggested IL-33 plays a critical role in microvascular homeostasis following barrier cell injury. KEY POINTS: Interleukin (IL)-33 causes a concentration-dependent reduction in feed artery diameter. IL-33 acts on vascular smooth muscle cells to augment Ca2+-mediated processes. IL-33 causes inhibitory phosphorylation of endothelial nitric oxide synthase and opposes endothelium-dependent dilatation. Engineered nanomaterial-induced lung injury and endothelial cell rupture in part act through IL-33 to mediate increased vascular tone.
Collapse
MESH Headings
- Animals
- Interleukin-33/metabolism
- Interleukin-33/pharmacology
- Rats, Sprague-Dawley
- Male
- Nitric Oxide Synthase Type III/metabolism
- Female
- Rats
- Vasodilation/drug effects
- Calcium/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiology
- Alarmins/metabolism
- Mesenteric Arteries/drug effects
- Mesenteric Arteries/physiology
- Arterioles/physiology
- Arterioles/drug effects
- Arterioles/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/physiology
- MAP Kinase Signaling System/drug effects
- MAP Kinase Signaling System/physiology
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiology
Collapse
Affiliation(s)
- Evan DeVallance
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Elizabeth Bowdridge
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Krista Garner
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Julie Griffith
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Madison Seman
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
| | - Thomas Batchelor
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Murugesan Velayutham
- Department of Biochemistry and Molecular Medicine, Health Sciences Center, West Virginia University, Morgantown, WV, USA
| | - W Travis Goldsmith
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Salik Hussain
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Eric E Kelley
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Timothy R Nurkiewicz
- Department of Physiology, Pharmacology & Toxicology, Health Sciences Center, West Virginia University, Morgantown, WV, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
2
|
Millen AE, Dighe S, Kordas K, Aminigo BZ, Zafron ML, Mu L. Air Pollution and Chronic Eye Disease in Adults: A Scoping Review. Ophthalmic Epidemiol 2024; 31:1-10. [PMID: 36864662 DOI: 10.1080/09286586.2023.2183513] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 02/18/2023] [Indexed: 03/04/2023]
Abstract
PURPOSE We conducted a scoping review of studies examining ambient air pollution as a risk factor for chronic eye disease influencing the lens, retina, and intraocular pressure in adults. METHODS Terms related to air pollution and eye disease outcomes were used to search for publications on Embase, Web of Science Core Collection, Global Health, PubMed, and the Cochrane Central Register of Controlled Trials from January 1, 2010, through April 11, 2022. RESULTS We identified 27 articles, focusing on the following non-mutually exclusive outcomes: cataract (n = 9), presbyopia (n = 1), retinal vein occlusion or central retinal arteriolar and venular equivalents (n = 5), intraocular pressure (IOP) (n = 3), glaucoma (n = 5), age-related macular degeneration (AMD) (n = 5), diabetic retinopathy (n = 2), and measures of retinal morphology (n = 3). Study designs included cross-sectional (n = 16), case-control (n = 4), and longitudinal (n = 7). Air pollutants were measured in 50% and 95% of the studies on lens and retina or IOP, respectively, and these exposures were assigned to geographic locations. Most research was conducted in global regions with high exposure to air pollution. Consistent associations suggested a possibly increased risk of cataract and retina-associated chronic eye disease with increasing exposure to particulate matter (PM2.5-PM10), NO2, NOx, and SO2. Associations with O3 were less consistent. CONCLUSIONS Accumulating research suggests air pollution may be a modifiable risk factor for chronic eye diseases of the lens and retina. The number of studies on each specific lens- or retina-related outcome is limited. Guidelines regarding the role of air pollution in chronic eye disease do not exist.
Collapse
Affiliation(s)
- Amy E Millen
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York, USA
| | - Shruti Dighe
- Department of Family Medicine, Allegheny Health Network Saint Vincent, Erie, PA, USA
| | - Katarzyna Kordas
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York, USA
| | - Boma Zelma Aminigo
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York, USA
| | - Michelle L Zafron
- Health Sciences at Abbott Library, University Libraries, University at Buffalo, Buffalo, New York, USA
| | - Lina Mu
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
3
|
Trembley JH, So SW, Nixon JP, Bowdridge EC, Garner KL, Griffith J, Engles KJ, Batchelor TP, Goldsmith WT, Tomáška JM, Hussain S, Nurkiewicz TR, Butterick TA. Whole-body inhalation of nano-sized carbon black: a surrogate model of military burn pit exposure. BMC Res Notes 2022; 15:275. [PMID: 35953874 PMCID: PMC9373276 DOI: 10.1186/s13104-022-06165-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 07/27/2022] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Chronic multisymptom illness (CMI) is an idiopathic disease affecting thousands of U.S. Veterans exposed to open-air burn pits emitting aerosolized particulate matter (PM) while serving in Central and Southwest Asia and Africa. Exposure to burn pit PM can result in profound biologic consequences including chronic fatigue, impaired cognition, and respiratory diseases. Dysregulated or unresolved inflammation is a possible underlying mechanism for CMI onset. We describe a rat model of whole-body inhalation exposure using carbon black nanoparticles (CB) as a surrogate for military burn pit-related exposure. Using this model, we measured biomarkers of inflammation in multiple tissues. RESULTS Male Sprague Dawley rats were exposed to CB aerosols by whole body inhalation (6 ± 0.83 mg/m3). Proinflammatory biomarkers were measured in multiple tissues including arteries, brain, lung, and plasma. Biomarkers of cardiovascular injury were also assayed in plasma. CB inhalation exposure increased CMI-related proinflammatory biomarkers such as IFN-γ and TNFα in multiple tissue samples. CB exposure also induced cardiovascular injury markers (adiponectin, MCP1, sE-Selectin, sICam-1 and TIMP1) in plasma. These findings support the validity of our animal exposure model for studies of burn pit-induced CMI. Future studies will model more complex toxicant mixtures as documented at multiple burn pit sites.
Collapse
Affiliation(s)
- Janeen H Trembley
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Simon W So
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, USA
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Joshua P Nixon
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, USA
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
- Burn Pits 360 Veterans Organization, Robstown, TX, USA
| | - Elizabeth C Bowdridge
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| | - Krista L Garner
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| | - Julie Griffith
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| | - Kevin J Engles
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| | - Thomas P Batchelor
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| | - William T Goldsmith
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| | | | - Salik Hussain
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), 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
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| | - Tammy A Butterick
- Minneapolis Veterans Affairs Health Care System, Minneapolis, MN, USA.
- Department of Food Science and Nutrition, University of Minnesota, St Paul, MN, USA.
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA.
- Center for Veterans Research and Education, Minneapolis, MN, USA.
| |
Collapse
|
4
|
Griffith JA, Garner KL, Bowdridge EC, DeVallance E, Schafner KJ, Engles KJ, Batchelor TP, Goldsmith WT, Wix K, Hussain S, Nurkiewicz TR. Nanomaterial Inhalation During Pregnancy Alters Systemic Vascular Function in a Cyclooxygenase-Dependent Manner. Toxicol Sci 2022; 188:219-233. [PMID: 35642938 PMCID: PMC9333412 DOI: 10.1093/toxsci/kfac055] [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: 11/14/2022] Open
Abstract
Pregnancy requires rapid adaptations in the uterine microcirculation to support fetal development. Nanomaterial inhalation is associated with cardiovascular dysfunction, which may impair gestation. We have shown that maternal nano-titanium dioxide (nano-TiO2) inhalation impairs microvascular endothelial function in response to arachidonic acid and thromboxane (TXA2) mimetics. However, the mechanisms underpinning this process are unknown. Therefore, we hypothesize that maternal nano-TiO2 inhalation during gestation results in uterine microvascular prostacyclin (PGI2) and TXA2 dysfunction. Pregnant Sprague-Dawley rats were exposed from gestational day 10-19 to nano-TiO2 aerosols (12.17 ± 1.67 mg/m3) or filtered air (sham-control). Dams were euthanized on gestational day 20, and serum, uterine radial arterioles, implantation sites, and lungs were collected. Serum was assessed for PGI2 and TXA2 metabolites. TXB2, the stable TXA2 metabolite, was significantly decreased in nano-TiO2 exposed dams (597.3 ± 84.4 vs 667.6 ± 45.6 pg/ml), whereas no difference was observed for 6-keto-PGF1α, the stable PGI2 metabolite. Radial arteriole pressure myography revealed that nano-TiO2 exposure caused increased vasoconstriction to the TXA2 mimetic, U46619, compared with sham-controls (-41.3% ± 4.3% vs -16.8% ± 3.4%). Nano-TiO2 exposure diminished endothelium-dependent vasodilation to carbaprostacyclin, a PGI2 receptor agonist, compared with sham-controls (30.0% ± 9.0% vs 53.7% ± 6.0%). Maternal nano-TiO2 inhalation during gestation decreased nano-TiO2 female pup weight when compared with sham-control males (3.633 ± 0.064 vs 3.995 ± 0.124 g). Augmented TXA2 vasoconstriction and decreased PGI2 vasodilation may lead to decreased placental blood flow and compromise maternofetal exchange of waste and nutrients, which could ultimately impact fetal health outcomes.
Collapse
Affiliation(s)
- Julie A Griffith
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
| | - Krista L Garner
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
| | - Elizabeth C Bowdridge
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
| | - Evan DeVallance
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
| | - Kallie J Schafner
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
| | - Kevin J Engles
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
| | - Thomas P Batchelor
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
| | - William T Goldsmith
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
| | - Kimberley Wix
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
| | - Salik Hussain
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
| | - Timothy R Nurkiewicz
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
- Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, West Virginia 26505-9229, USA
| |
Collapse
|
5
|
Alsaleh NB. Adverse cardiovascular responses of engineered nanomaterials: Current understanding of molecular mechanisms and future challenges. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 37:102421. [PMID: 34166839 DOI: 10.1016/j.nano.2021.102421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 04/14/2021] [Accepted: 05/09/2021] [Indexed: 11/30/2022]
Abstract
Nanotechnology is spanning multiple fields of study from materials science to computer engineering and drug discovery. Since the early 21st century, nanotechnology and nano-enabled research have received great attention and governmental funding accompanied with interest to ensure human and environmental safety of engineered nanomaterials (ENMs). Optimal functioning of the cardiovascular (CV) system is of utmost importance for the overall health of the body. Following exposure, ENMs essentially end up in the circulation (at least partially) and hence it is key to assess any associated adverse CV consequences. Accumulating research suggests that exposure to ENMs (different compositions and physicochemical properties) has the capacity to directly and indirectly interact with CV components resulting in adverse events and worsening of CV complications. However, the underlying molecular mechanisms driving these events remain to be elucidated. In this article, we review state-of-art literature on ENM-associated adverse CV responses and discuss the potential underlying molecular mechanisms.
Collapse
Affiliation(s)
- Nasser B Alsaleh
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; Nanobiotechnology Unit, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
6
|
Mohamadzadeh N, Zirak Javanmard M, Karimipour M, Farjah G. Developmental Toxicity of the Neural Tube Induced by Titanium Dioxide Nanoparticles in Mouse Embryos. Avicenna J Med Biotechnol 2021; 13:74-80. [PMID: 34012522 PMCID: PMC8112145 DOI: 10.18502/ajmb.v13i2.5524] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Background: This study investigated the potential effects of Titanium dioxide nanoparticles (Tio2NPs) followed by maternal gavage on fetal development and neural tube formation during pregnancy in mice. Methods: Thirty pregnant mice were randomly divided into five main study groups including the untreated control and 4 experimental groups (n=6 per group). The control group was treated with normal saline and the experimental groups were orally treated with doses of 30, 150, 300, and 500 mg/kg Body Weight (BW) of Tio2NPs during pregnancy. On gestational day 16 and 19 (n=3 per group), pregnant mice were euthanized and then examined for neural tube defects and compared with control. Serial transverse sections were prepared in both cranial region and in lumbar region of spinal cord. Results: Treatment with Tio2NPs resulted in low fetal weight and short length, dilation of lateral ventricle, thinning of cerebral cortex and spinal cord, spina bifida occulta and an increase in the number of apoptotic neurons in exposed embryos at doses of 300 and 500 mg/kg (p<0.05). Conclusion: It seems that exposure to nanoparticles of Tio2 during pregnancy induces growth retardation and for the first time, teratogenicity of this nanomaterial in neural tube development and induction of defects such as spinal bifida, reduction in cortical thickness and dilatation of lateral ventricles were verified which can be related to incidence of apoptosis in central nervous system.
Collapse
Affiliation(s)
- Nahid Mohamadzadeh
- Department of Anatomical Sciences, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Masoumeh Zirak Javanmard
- Department of Anatomical Sciences, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mojtaba Karimipour
- Department of Anatomical Sciences, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Gholamhosain Farjah
- Department of Anatomical Sciences, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
7
|
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
|
8
|
Dréno B, Alexis A, Chuberre B, Marinovich M. Safety of titanium dioxide nanoparticles in cosmetics. J Eur Acad Dermatol Venereol 2019; 33 Suppl 7:34-46. [DOI: 10.1111/jdv.15943] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/03/2019] [Indexed: 12/31/2022]
Affiliation(s)
- B. Dréno
- Onco‐Dermatology Department CHU Nantes CRCINA University Nantes Nantes France
| | - A. Alexis
- Department of Dermatology Icahn School of Medicine at Mount Sinai New York NY USA
| | - B. Chuberre
- L'Oréal Cosmetique Active International Levallois‐Perret France
| | - M. Marinovich
- Department of Pharmacological and Biomolecular Sciences University of Milan Milan Italy
| |
Collapse
|
9
|
Redox interactions and genotoxicity of metal-based nanoparticles: A comprehensive review. Chem Biol Interact 2019; 312:108814. [PMID: 31509734 DOI: 10.1016/j.cbi.2019.108814] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 08/11/2019] [Accepted: 09/05/2019] [Indexed: 12/25/2022]
Abstract
Nanotechnology is a growing science that may provide several new applications for medicine, food preservation, diagnostic technologies, and sanitation. Despite its beneficial applications, there are several questions related to the safety of nanomaterials for human use. The development of nanotechnology is associated with some concerns because of the increased risk of carcinogenesis following exposure to nanomaterials. The increased levels of reactive oxygen species (ROS) that are due to exposure to nanoparticles (NPs) are primarily responsible for the genotoxicity of metal NPs. Not all, but most metal NPs are able to directly produce free radicals through the release of metal ions and through interactions with water molecules. Furthermore, the increased production of free radicals and the cell death caused by metal NPs can stimulate reduction/oxidation (redox) reactions, leading to the continuous endogenous production of ROS in a positive feedback loop. The overexpression of inflammatory mediators, such as NF-kB and STATs, the mitochondrial malfunction and the increased intracellular calcium levels mediate the chronic oxidative stress that occurs after exposure to metal NPs. In this paper, we review the genotoxicity of different types of metal NPs and the redox mechanisms that amplify the toxicity of these NPs.
Collapse
|
10
|
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
|
11
|
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
|
12
|
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
|
13
|
Vasomotor dysfunction in human subcutaneous arteries exposed ex vivo to food-grade titanium dioxide. Food Chem Toxicol 2018; 120:321-327. [DOI: 10.1016/j.fct.2018.07.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/04/2018] [Accepted: 07/06/2018] [Indexed: 01/22/2023]
|
14
|
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
|
15
|
Cao Y, Gong Y, Liao W, Luo Y, Wu C, Wang M, Yang Q. A review of cardiovascular toxicity of TiO 2, ZnO and Ag nanoparticles (NPs). Biometals 2018; 31:457-476. [PMID: 29748744 DOI: 10.1007/s10534-018-0113-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 05/04/2018] [Indexed: 12/22/2022]
Abstract
To ensure the safe use of nanoparticles (NPs) in modern society, it is necessary and urgent to assess the potential toxicity of NPs. Cardiovascular system is required for the systemic distribution of NPs entering circulation. Therefore, the adverse cardiovascular effects of NPs have gained extensive research interests. Metal based NPs, such as TiO2, ZnO and Ag NPs, are among the most popular NPs found in commercially available products. They may also have potential applications in biomedicine, which could increase their contact with cardiovascular systems. This review aimed at providing an overview about the adverse cardiovascular effects of TiO2, ZnO and Ag NPs. We discussed about the bio-distribution of NPs following different exposure routes. We also discussed about the cardiovascular toxicity of TiO2, ZnO and Ag NPs as assessed by in vivo and in vitro models. The possible mechanisms and contribution of physicochemical properties of metal based NPs were also discussed.
Collapse
Affiliation(s)
- Yi Cao
- Key Laboratory of Environment-Friendly Chemistry and Application of Ministry of Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, 411105, People's Republic of China.
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, No. 1023 South Shatai Road, Guangzhou, 510515, People's Republic of China.
| | - Yu Gong
- Key Laboratory of Environment-Friendly Chemistry and Application of Ministry of Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, 411105, People's Republic of China
| | - Wenzhen Liao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, No. 1023 South Shatai Road, Guangzhou, 510515, People's Republic of China.
| | - Yunfeng Luo
- Key Laboratory of Environment-Friendly Chemistry and Application of Ministry of Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, 411105, People's Republic of China
| | - Chaohua Wu
- Key Laboratory of Environment-Friendly Chemistry and Application of Ministry of Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, 411105, People's Republic of China
| | - Maolin Wang
- Key Laboratory of Environment-Friendly Chemistry and Application of Ministry of Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, 411105, People's Republic of China
| | - Qianyu Yang
- Key Laboratory of Environment-Friendly Chemistry and Application of Ministry of Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, 411105, People's Republic of China
| |
Collapse
|
16
|
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
|
17
|
Vasomotor function in rat arteries after ex vivo and intragastric exposure to food-grade titanium dioxide and vegetable carbon particles. Part Fibre Toxicol 2018; 15:12. [PMID: 29482579 PMCID: PMC5828140 DOI: 10.1186/s12989-018-0248-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 02/07/2018] [Indexed: 02/07/2023] Open
Abstract
Background Humans are continuously exposed to particles in the gastrointestinal tract. Exposure may occur directly through ingestion of particles via food or indirectly by removal of inhaled material from the airways by the mucociliary clearance system. We examined the effects of food-grade particle exposure on vasomotor function and systemic oxidative stress in an ex vivo study and intragastrically exposed rats. Methods In an ex vivo study, aorta rings from naïve Sprague-Dawley rats were exposed for 30 min to food-grade TiO2 (E171), benchmark TiO2 (Aeroxide P25), food-grade vegetable carbon (E153) or benchmark carbon black (Printex 90). Subsequently, the vasomotor function was assessed in wire myographs. In an in vivo study, lean Zucker rats were exposed intragastrically once a week for 10 weeks to vehicle, E171 or E153. Doses were comparable to human daily intake. Vasomotor function in the coronary arteries and aorta was assessed using wire myographs. Tetrahydrobiopterin, ascorbate, malondialdehyde and asymmetric dimethylarginine were measured in blood as markers of oxidative stress and vascular function. Results Direct exposure of E171 to aorta rings ex vivo increased the acetylcholine-induced vasorelaxation and 5-hydroxytryptamine-induced vasocontraction. E153 only increased acetylcholine-induced vasorelaxation, and Printex 90 increased the 5-hydroxytryptamine-induced vasocontraction, whereas Aeroxide P25 did not affect the vasomotor function. In vivo exposure showed similar results as ex vivo exposure; increased acetylcholine-induced vasorelaxation in coronary artery segments of E153 and E171 exposed rats, whereas E171 exposure altered 5-hydroxytryptamine-induced vasocontraction in distal coronary artery segments. Plasma levels of markers of oxidative stress and vascular function showed no differences between groups. Conclusion Gastrointestinal tract exposure to E171 and E153 was associated with modest albeit statistically significant alterations in the vasocontraction and vasorelaxation responses. Direct particle exposure to aorta rings elicited a similar type of response. The vasomotor responses were not related to biomarkers of systemic oxidative stress.
Collapse
|
18
|
In Vivo Protective Effects of Nootkatone against Particles-Induced Lung Injury Caused by Diesel Exhaust Is Mediated via the NF-κB Pathway. Nutrients 2018; 10:nu10030263. [PMID: 29495362 PMCID: PMC5872681 DOI: 10.3390/nu10030263] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 01/18/2018] [Accepted: 01/23/2018] [Indexed: 12/21/2022] Open
Abstract
Numerous studies have shown that acute particulate air pollution exposure is linked with pulmonary adverse effects, including alterations of pulmonary function, inflammation, and oxidative stress. Nootkatone, a constituent of grapefruit, has antioxidant and anti-inflammatory effects. However, the effect of nootkatone on lung toxicity has not been reported so far. In this study we evaluated the possible protective effects of nootkatone on diesel exhaust particles (DEP)-induced lung toxicity, and the possible mechanisms underlying these effects. Mice were intratracheally (i.t.) instilled with either DEP (30 µg/mouse) or saline (control). Nootkatone was given to mice by gavage, 1 h before i.t. instillation, with either DEP or saline. Twenty-four hours following DEP exposure, several physiological and biochemical endpoints were assessed. Nootkatone pretreatment significantly prevented the DEP-induced increase in airway resistance in vivo, decreased neutrophil infiltration in bronchoalveolar lavage fluid, and abated macrophage and neutrophil infiltration in the lung interstitium, assessed by histolopathology. Moreover, DEP caused a significant increase in lung concentrations of 8-isoprostane and tumor necrosis factor α, and decreased the reduced glutathione concentration and total nitric oxide activity. These actions were all significantly alleviated by nootkatone pretreatment. Similarly, nootkatone prevented DEP-induced DNA damage and prevented the proteolytic cleavage of caspase-3. Moreover, nootkatone inhibited nuclear factor-kappaB (NF-κB) induced by DEP. We conclude that nootkatone prevented the DEP-induced increase in airway resistance, lung inflammation, oxidative stress, and the subsequent DNA damage and apoptosis through a mechanism involving inhibition of NF-κB activation. Nootkatone could possibly be considered a beneficial protective agent against air pollution-induced respiratory adverse effects.
Collapse
|
19
|
Giles LV, Tebbutt SJ, Carlsten C, Koehle MS. The effect of low and high-intensity cycling in diesel exhaust on flow-mediated dilation, circulating NOx, endothelin-1 and blood pressure. PLoS One 2018; 13:e0192419. [PMID: 29466393 PMCID: PMC5821322 DOI: 10.1371/journal.pone.0192419] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 01/11/2018] [Indexed: 01/12/2023] Open
Abstract
INTRODUCTION Exposure to air pollution impairs aspects of endothelial function such as flow-mediated dilation (FMD). Outdoor exercisers are frequently exposed to air pollution, but how exercising in air pollution affects endothelial function and how these effects are modified by exercise intensity are poorly understood. OBJECTIVES Therefore, the purpose of this study was to determine the effects of low-intensity and high-intensity cycling with diesel exhaust (DE) exposure on FMD, blood pressure, plasma nitrite and nitrate (NOx) and endothelin-1. METHODS Eighteen males performed 30-minute trials of low or high-intensity cycling (30% and 60% of power at VO2peak) or a resting control condition. For each subject, each trial was performed once while breathing filtered air (FA) and once while breathing DE (300ug/m3 of PM2.5, six trials in total). Preceding exposure, immediately post-exposure, 1 hour and 2 hours post-exposure, FMD, blood pressure and plasma endothelin-1 and NOx concentrations were measured. Data were analyzed using repeated-measures ANOVA and linear mixed model. RESULTS Following exercise in DE, plasma NOx significantly increased and was significantly greater than FA (p<0.05). Two hours following DE exposure, endothelin-1 was significantly less than FA (p = 0.037) but exercise intensity did not modify this response. DE exposure did not affect FMD or blood pressure. CONCLUSION Our results suggest that exercising in DE did not adversely affect plasma NOX, endothelin-1, FMD and blood pressure. Therefore, recommendations for healthy individuals to moderate or avoid exercise during bouts of high pollution appear to have no acute protective effect.
Collapse
Affiliation(s)
- Luisa V. Giles
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Scott J. Tebbutt
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Heart Lung Innovation, University of British Columbia and St. Paul’s Hospital, Vancouver, British Columbia, Canada
| | - Christopher Carlsten
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Heart Lung Innovation, University of British Columbia and St. Paul’s Hospital, Vancouver, British Columbia, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael S. Koehle
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
- Division of Sports Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
20
|
Nichols CE, Shepherd DL, Hathaway QA, Durr AJ, Thapa D, Abukabda A, Yi J, Nurkiewicz TR, Hollander JM. Reactive oxygen species damage drives cardiac and mitochondrial dysfunction following acute nano-titanium dioxide inhalation exposure. Nanotoxicology 2018; 12:32-48. [PMID: 29243970 PMCID: PMC5777890 DOI: 10.1080/17435390.2017.1416202] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 12/07/2017] [Accepted: 12/07/2017] [Indexed: 12/25/2022]
Abstract
Nanotechnology offers innovation in products from cosmetics to drug delivery, leading to increased engineered nanomaterial (ENM) exposure. Unfortunately, health impacts of ENM are not fully realized. Titanium dioxide (TiO2) is among the most widely produced ENM due to its use in numerous applications. Extrapulmonary effects following pulmonary exposure have been identified and may involve reactive oxygen species (ROS). The goal of this study was to determine the extent of ROS involvement on cardiac function and the mitochondrion following nano-TiO2 exposure. To address this question, we utilized a transgenic mouse model with overexpression of a novel mitochondrially-targeted antioxidant enzyme (phospholipid hydroperoxide glutathione peroxidase; mPHGPx) which provides protection against oxidative stress to lipid membranes. MPHGPx mice and littermate controls were exposed to nano-TiO2 aerosols (Evonik, P25) to provide a calculated pulmonary deposition of 11 µg/mouse. Twenty-four hours following exposure, we observed diastolic dysfunction as evidenced by E/A ratios greater than 2 and increased radial strain during diastole in wild-type mice (p < 0.05 for both), indicative of restrictive filling. Overexpression of mPHGPx mitigated the contractile deficits resulting from nano-TiO2 exposure. To investigate the cellular mechanisms associated with the observed cardiac dysfunction, we focused our attention on the mitochondrion. We observed a significant increase in ROS production (p < 0.05) and decreased mitochondrial respiratory function (p < 0.05) following nano-TiO2 exposure which were attenuated in mPHGPx transgenic mice. In summary, nano-TiO2 inhalation exposure is associated with cardiac diastolic dysfunction and mitochondrial functional alterations, which can be mitigated by the overexpression of mPHGPx, suggesting ROS contribution in the development of contractile and bioenergetic dysfunction.
Collapse
Affiliation(s)
- Cody E. Nichols
- Division of Exercise Physiology; West Virginia University School of Medicine, Morgantown, WV 26506
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, United States
| | - Danielle L. Shepherd
- Division of Exercise Physiology; West Virginia University School of Medicine, Morgantown, WV 26506
- Mitochondria, Metabolism & Bioenergetics Working Group; West Virginia University School of Medicine, Morgantown, WV 26506
| | - Quincy A. Hathaway
- Division of Exercise Physiology; West Virginia University School of Medicine, Morgantown, WV 26506
- Mitochondria, Metabolism & Bioenergetics Working Group; West Virginia University School of Medicine, Morgantown, WV 26506
| | - Andrya J. Durr
- Division of Exercise Physiology; West Virginia University School of Medicine, Morgantown, WV 26506
- Mitochondria, Metabolism & Bioenergetics Working Group; West Virginia University School of Medicine, Morgantown, WV 26506
| | - Dharendra Thapa
- Division of Exercise Physiology; West Virginia University School of Medicine, Morgantown, WV 26506
| | - Alaeddin Abukabda
- Department of Physiology and Pharmacology; West Virginia University School of Medicine, Morgantown, WV 26506
| | - Jinghai Yi
- Department of Physiology and Pharmacology; West Virginia University School of Medicine, Morgantown, WV 26506
| | - Timothy R. Nurkiewicz
- Mitochondria, Metabolism & Bioenergetics Working Group; West Virginia University School of Medicine, Morgantown, WV 26506
- Department of Physiology and Pharmacology; West Virginia University School of Medicine, Morgantown, WV 26506
| | - John M. Hollander
- Division of Exercise Physiology; West Virginia University School of Medicine, Morgantown, WV 26506
- Mitochondria, Metabolism & Bioenergetics Working Group; West Virginia University School of Medicine, Morgantown, WV 26506
| |
Collapse
|
21
|
Vidanapathirana AK, Thompson LC, Herco M, Odom J, Sumner SJ, Fennell TR, Brown JM, Wingard CJ. Acute intravenous exposure to silver nanoparticles during pregnancy induces particle size and vehicle dependent changes in vascular tissue contractility in Sprague Dawley rats. Reprod Toxicol 2018; 75:10-22. [PMID: 29154916 PMCID: PMC6241519 DOI: 10.1016/j.reprotox.2017.11.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 10/26/2017] [Accepted: 11/13/2017] [Indexed: 12/17/2022]
Abstract
The use of silver nanoparticles (AgNP) raises safety concerns during susceptible life stages such as pregnancy. We hypothesized that acute intravenous exposure to AgNP during late stages of pregnancy will increase vascular tissue contractility, potentially contributing to alterations in fetal growth. Sprague Dawley rats were exposed to a single dose of PVP or Citrate stabilized 20 or 110nm AgNP (700μg/kg). Differential vascular responses and EC50 values were observed in myographic studies in uterine, mesenteric arteries and thoracic aortic segments, 24h post-exposure. Reciprocal responses were observed in aortic and uterine vessels following PVP stabilized AgNP with an increased force of contraction in uterine artery and increased relaxation responses in aorta. Citrate stabilized AgNP exposure increased contractile force in both uterine and aortic vessels. Intravenous AgNP exposure during pregnancy displayed particle size and vehicle dependent moderate changes in vascular tissue contractility, potentially influencing fetal blood supply.
Collapse
Affiliation(s)
- A K Vidanapathirana
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - L C Thompson
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - M Herco
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - J Odom
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - S J Sumner
- Discovery Sciences, RTI International, Research Triangle Park, NC, 27709, USA; Department of Nutrition School of Public Health University of North Carolina at Chapel Hill, Kannapolis, NC, 28081, USA
| | - T R Fennell
- Discovery Sciences, RTI International, Research Triangle Park, NC, 27709, USA
| | - J M Brown
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, CO, 80045, USA
| | - C J Wingard
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA; Department of Physical Therapy, Bellarmine University, Louisville, KY, 40205, USA.
| |
Collapse
|
22
|
Mohamed T, Matou-Nasri S, Farooq A, Whitehead D, Azzawi M. Polyvinylpyrrolidone-coated gold nanoparticles inhibit endothelial cell viability, proliferation, and ERK1/2 phosphorylation and reduce the magnitude of endothelial-independent dilator responses in isolated aortic vessels. Int J Nanomedicine 2017; 12:8813-8830. [PMID: 29263670 PMCID: PMC5732551 DOI: 10.2147/ijn.s133093] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Background Gold nanoparticles (AuNPs) demonstrate clinical potential for drug delivery and imaging diagnostics. As AuNPs aggregate in physiological fluids, polymer-surface modifications are utilized to allow their stabilization and enhance their retention time in blood. However, the impact of AuNPs on blood vessel function remains poorly understood. In the present study, we investigated the effects of AuNPs and their stabilizers on endothelial cell (EC) and vasodilator function. Materials and methods Citrate-stabilized AuNPs (12±3 nm) were synthesized and surface-modified using mercapto polyethylene glycol (mPEG) and polyvinylpyrrolidone (PVP) polymers. Their uptake by isolated ECs and whole vessels was visualized using transmission electron microscopy and quantified using inductively coupled plasma mass spectrometry. Their biological effects on EC proliferation, viability, apoptosis, and the ERK1/2-signaling pathway were determined using automated cell counting, flow cytometry, and Western blotting, respectively. Endothelial-dependent and independent vasodilator functions were assessed using isolated murine aortic vessel rings ex vivo. Results AuNPs were located in endothelial endosomes within 30 minutes’ exposure, while their surface modification delayed this cellular uptake over time. After 24 hours’ exposure, all AuNPs (including polymer-modified AuNPs) induced apoptosis and decreased cell viability/proliferation. These inhibitory effects were lost after 48 hours’ exposure (except for the PVP-modified AuNPs). Furthermore, all AuNPs decreased acetylcholine (ACh)-induced phosphorylation of ERK1/2, a key signaling protein of cell function. mPEG-modified AuNPs had lower cytostatic effects than PVP-modified AuNPs. Citrate-stabilized AuNPs did not alter endothelial-dependent vasodilation induced by ACh, but attenuated endothelial-independent responses induced by sodium nitroprusside. PVP-modified AuNPs attenuated ACh-induced dilation, whereas mPEG-modified AuNPs did not, though this was dose-related. Conclusion We demonstrated that mPEG-modified AuNPs at a therapeutic dosage showed lower cytostatic effects and were less detrimental to vasodilator function than PVP-modified AuNPs, indicating greater potential as agents for diagnostic imaging and therapy.
Collapse
Affiliation(s)
- Teba Mohamed
- School of Healthcare Science, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK
| | - Sabine Matou-Nasri
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Centre, National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Asima Farooq
- School of Science and the Environment, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK
| | - Debra Whitehead
- School of Science and the Environment, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK
| | - May Azzawi
- School of Healthcare Science, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK
| |
Collapse
|
23
|
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
|
24
|
Kang LS, Masilamani S, Boegehold MA. Juvenile growth reduces the influence of epithelial sodium channels on myogenic tone in skeletal muscle arterioles. Clin Exp Pharmacol Physiol 2017; 43:1199-1207. [PMID: 27560463 DOI: 10.1111/1440-1681.12664] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 06/21/2016] [Accepted: 07/14/2016] [Indexed: 02/05/2023]
Abstract
Previous studies have documented that rapid juvenile growth is accompanied by functional changes in the arteriolar endothelium, but much less is known about functional changes in arteriolar smooth muscle over this period. In this study, we investigate the possible contribution of epithelial sodium channels (ENaC) to the myogenic behaviour of arterioles at two stages of juvenile growth. The effects of the ENaC inhibitor benzamil on different levels of myogenic tone were studied in isolated gracilis muscle arterioles from rats aged 21-28 days ("weanlings") and 42-49 days ("juveniles"). ENaC subunit expression in the arteriolar wall was also determined, and the interaction between ENaC and nitric oxide (NO) in regulating vascular tone was explored by combined use of benzamil and NG -monomethyl-l-arginine (l-NMMA). At physiological pressures, both steady-state myogenic tone and the dynamic adjustments in this tone triggered by acute pressure changes were less in juvenile arterioles than in weanling arterioles. α, β and γ ENaC protein was present in arterioles at both ages, but benzamil only had an effect on myogenic tone in weanling arterioles. In these vessels, benzamil increased, rather than decreased, myogenic tone, and this effect was prevented by l-NMMA or endothelial removal. These findings suggest that although ENaC is present in gracilis muscle arterioles of both weanling and juvenile rats, it is not obligatory for the genesis of myogenic activity in these vessels at either age. However, ENaC activity can significantly modulate the level of myogenic tone through stimulation of endothelial NO release at an early stage of growth.
Collapse
Affiliation(s)
- Lori S Kang
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Shyama Masilamani
- Department of Internal Medicine/Division of Nephrology, Virginia Commonwealth University Medical Center, Richmond, VA, USA
| | - Matthew A Boegehold
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
25
|
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
|
26
|
Hong F, Yu X, Wu N, Zhang YQ. Progress of in vivo studies on the systemic toxicities induced by titanium dioxide nanoparticles. Toxicol Res (Camb) 2017; 6:115-133. [PMID: 30090482 PMCID: PMC6061230 DOI: 10.1039/c6tx00338a] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 12/09/2016] [Indexed: 01/29/2023] Open
Abstract
Titanium dioxide nanoparticles (TiO2 NPs) are inorganic materials with a diameter of 1-100 nm. In recent years, TiO2 NPs have been used in a wide range of products, including food, toothpaste, cosmetics, medicine, paints and printing materials, due to their unique properties (high stability, anti-corrosion, and efficient photocatalysis). Following exposure via various routes including inhalation, injection, dermal deposition and gastrointestinal tract absorption, NPs can be found in various organs in the body potentially inducing toxic effects. Thus more attention to the safety of TiO2 NPs is necessary. Therefore, the present review aims to provide a comprehensive evaluation of the toxic effects induced by TiO2 NPs in the lung, liver, stomach, intestine, kidney, spleen, brain, hippocampus, heart, blood vessels, ovary and testis of mice and rats in in vivo experiments, and evaluate their potential toxic mechanisms. The findings will provide an important reference for human risk evaluation and management following TiO2 NP exposure.
Collapse
Affiliation(s)
- Fashui Hong
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection , Huaiyin Normal University , Huaian 223300 , China .
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake , Huaiyin Normal University , Huaian 223300 , China
- School of Life Sciences , Huaiyin Normal University , Huaian 223300 , China
| | - Xiaohong Yu
- School of Basic Medical and Biological Sciences , Soochow University , Suzhou 215123 , China .
| | - Nan Wu
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection , Huaiyin Normal University , Huaian 223300 , China .
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake , Huaiyin Normal University , Huaian 223300 , China
- School of Life Sciences , Huaiyin Normal University , Huaian 223300 , China
| | - Yu-Qing Zhang
- School of Basic Medical and Biological Sciences , Soochow University , Suzhou 215123 , China .
| |
Collapse
|
27
|
Tallon LA, Manjourides J, Pun VC, Mittleman MA, Kioumourtzoglou MA, Coull B, Suh H. Erectile dysfunction and exposure to ambient Air pollution in a nationally representative cohort of older Men. Environ Health 2017; 16:12. [PMID: 28212639 PMCID: PMC5316194 DOI: 10.1186/s12940-017-0216-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 02/08/2017] [Indexed: 05/29/2023]
Abstract
BACKGROUND Little is known about the association between air pollution and erectile dysfunction (ED), a disorder occurring in 64% of men over the age of 70, and to date, no studies have been published. To address this significant knowledge gap, we explored the relationship between ED and air pollution in a group of older men who were part of the National Social Life, Health, and Aging Project (NSHAP), a nationally representative cohort study of older Americans. METHODS We obtained incident ED status and participant data for 412 men (age 57-85). Fine particulate matter (PM2.5) exposures were estimated using spatio-temporal models based on participants' geocoded addresses, while nitrogen dioxide (NO2) and ozone (O3) concentrations were estimated using nearest measurements from the Environmental Protection Agency's Air Quality System. The association between air pollution and incident ED (newly developed in Wave 2) was examined and logistic regression models were run with adjusted models controlling for race, education, season, smoking, obesity, diabetes, depression, and median household income of census tract. RESULTS We found positive, although statistically insignificant, associations between PM2.5, NO2, and O3 exposures and odds of incident ED for each of our examined exposure windows, including 1 to 7 year moving averages. Odds ratios (OR) for 1 and 7 year moving averages equaled 1.16 (95% CI: 0.87, 1.55) and 1.16 (95% CI: 0.92, 1.46), respectively, for an IQR increase in PM2.5 exposures. Observed associations were robust to model specifications and were not significantly modified by any of the examined risk factors for ED. CONCLUSIONS We found associations between PM2.5, NO2, and O3 exposures and odds of developing ED that did not reach nominal statistical significance, although exposures to each pollutant were consistently associated with higher odds of developing ED. While more research is needed, our findings suggest a relationship between air pollutant exposure and incident cases of ED, a common condition in older men.
Collapse
Affiliation(s)
- Lindsay A. Tallon
- Department of Health Sciences, Northeastern University, 360 Huntington Avenue, Boston, MA 02115 USA
- MCPHS University, 179 Longwood Avenue, Boston, MA 02115 USA
| | - Justin Manjourides
- Department of Health Sciences, Northeastern University, 360 Huntington Avenue, Boston, MA 02115 USA
| | - Vivian C. Pun
- Department of Health Sciences, Northeastern University, 360 Huntington Avenue, Boston, MA 02115 USA
| | - Murray A. Mittleman
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave., Boston, MA 02115 USA
| | - Marianthi-Anna Kioumourtzoglou
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, 722 W. 168th Street, #1105C, New York, NY 10032 USA
| | - Brent Coull
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Ave., Boston, MA 02115 USA
| | - Helen Suh
- Department of Civil and Environmental Engineering, Tufts University, 200 College Avenue, 301 Anderson Hall, Medford, MA 02155 USA
| |
Collapse
|
28
|
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
|
29
|
Abukabda AB, Stapleton PA, Nurkiewicz TR. Metal Nanomaterial Toxicity Variations Within the Vascular System. Curr Environ Health Rep 2016; 3:379-391. [PMID: 27686080 PMCID: PMC5112123 DOI: 10.1007/s40572-016-0112-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Engineered nanomaterials (ENM) are anthropogenic materials with at least one dimension less than 100 nm. Their ubiquitous employment in biomedical and industrial applications in the absence of full toxicological assessments raises significant concerns over their safety on human health. This is a significant concern, especially for metal and metal oxide ENM as they may possess the greatest potential to impair human health. A large body of literature has developed that reflects adverse systemic effects associated with exposure to these materials, but an integrated mechanistic framework for how ENM exposure influences morbidity remains elusive. This may be due in large part to the tremendous diversity of existing ENM and the rate at which novel ENM are produced. In this review, the influence of specific ENM physicochemical characteristics and hemodynamic factors on cardiovascular toxicity is discussed. Additionally, the toxicity of metallic and metal oxide ENM is presented in the context of the cardiovascular system and its discrete anatomical and functional components. Finally, future directions and understudied topics are presented. While it is clear that the nanotechnology boom has increased our interest in ENM toxicity, it is also evident that the field of cardiovascular nanotoxicology remains in its infancy and continued, expansive research is necessary in order to determine the mechanisms via which ENM exposure contributes to cardiovascular morbidity.
Collapse
Affiliation(s)
- Alaeddin B. Abukabda
- Center for Cardiovascular and Respiratory Sciences, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Phoebe A. Stapleton
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ, USA
| | - Timothy R. Nurkiewicz
- Center for Cardiovascular and Respiratory Sciences, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
30
|
Shukur A, Whitehead D, Seifalian A, Azzawi M. The influence of silica nanoparticles on small mesenteric arterial function. Nanomedicine (Lond) 2016; 11:2131-46. [DOI: 10.2217/nnm-2016-0124] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To determine the influence of silica nanoparticles (SiNPs) on small arterial function; both ex vivo and in vivo. Methods: Mono-dispersed dye-encapsulated SiNPs (97.85 ± 2.26 nm) were fabricated and vasoconstrictor and vasodilator responses of mesenteric arteries assessed. Results: We show that while exposure to SiNPs under static conditions, attenuated endothelial dependent dilator responses ex vivo, attenuation was only evident at lower agonist concentrations, when exposed under flow conditions or after intravenous administration in vivo. Pharmacological inhibition studies suggest that SiNPs may interfere with the endothelial dependent hyperpolarizing factor vasodilator pathway. Conclusion: The dosage dependent influence of SiNPs on arterial function will help identify strategies for their safe clinical administration.
Collapse
Affiliation(s)
- Ali Shukur
- School of Healthcare Science, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, UK
| | - Debra Whitehead
- School of Science & the Environment, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, UK
| | - Alexander Seifalian
- Centre for Nanotechnology & Regenerative Medicine, UCL Division of Surgery & Interventional Science, University College London, London, UK
| | - May Azzawi
- School of Healthcare Science, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, UK
| |
Collapse
|
31
|
Zhu X, Hou L, Zhang J, Yao C, Liu Y, Zhang C, Xu Y, Cao J. The structural and functional effects of fine particulate matter from cooking oil fumes on rat umbilical cord blood vessels. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2016; 23:16567-16578. [PMID: 27178289 DOI: 10.1007/s11356-016-6821-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 05/02/2016] [Indexed: 06/05/2023]
Abstract
A growing body of epidemiological evidence has supported the association between maternal exposure to airborne fine particulate matter (PM2.5) during pregnancy and adverse pregnancy outcomes. However, the specific biological mechanisms implicated in the causes of adverse pregnancy outcomes are not well defined. In this study, a pregnant rat model of exposure to different doses of cooking oil fumes (COFs)-derived PM2.5 by tail intravenous injection in different pregnant stages was established. The results indicated that exposure to COFs-derived PM2.5 was associated with adverse pregnancy outcomes, changed the structure of umbilical cord blood vessels, decreased the diameter and lumen area, and increased wall thickness. What's more, a significant increase of maximum contraction tension was observed in the early pregnancy high-dose exposure group and pregnant low-dose exposure group compared to the control group. Based on the maximum contraction tension, acetylcholine (ACh) did not induce vasodilation but caused a dose-dependent constriction, and there were significant differences in the two groups compared to the control group. Exposure to COFs-derived PM2.5 impaired the vasomotor function of umbilical veins by affecting the expression of NO and ET-1. This is the first study that evaluated the association of risk of adverse pregnancy outcomes and pregnant rats exposed to COFs-derived PM2.5 and primarily explored the potential mechanisms of umbilical cord blood vessels injury on a rat model. More detailed vitro and vivo studies are needed to further explore the mechanism in the future.
Collapse
Affiliation(s)
- Xiaoxia Zhu
- Department of Occupational and Environmental, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, China
| | - Lijuan Hou
- Department of Occupational and Environmental, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, China
| | - Jian Zhang
- Department of Occupational and Environmental, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, China
| | - Cijiang Yao
- Department of Occupational and Environmental, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, China
| | - Ying Liu
- Department of Occupational and Environmental, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, China
| | - Chao Zhang
- Department of Occupational and Environmental, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, China
| | - Yachun Xu
- Department of Occupational and Environmental, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, China
| | - Jiyu Cao
- The Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, Meishan Road 81, Hefei, 230032, Anhui, China.
| |
Collapse
|
32
|
Shakeel M, Jabeen F, Shabbir S, Asghar MS, Khan MS, Chaudhry AS. Toxicity of Nano-Titanium Dioxide (TiO2-NP) Through Various Routes of Exposure: a Review. Biol Trace Elem Res 2016; 172:1-36. [PMID: 26554951 DOI: 10.1007/s12011-015-0550-x] [Citation(s) in RCA: 186] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 10/19/2015] [Indexed: 01/18/2023]
Abstract
Nano-titanium dioxide (TiO2) is one of the most commonly used materials being synthesized for use as one of the top five nanoparticles. Due to the extensive application of TiO2 nanoparticles and their inclusion in many commercial products, the increased exposure of human beings to nanoparticles is possible. This exposure could be routed via dermal penetration, inhalation and oral ingestion or intravenous injection. Therefore, regular evaluation of their potential toxicity and distribution in the bodies of exposed individuals is essential. Keeping in view the potential health hazards of TiO2 nanoparticles for humans, we reviewed the research articles about studies performed on rats or other mammals as animal models. Most of these studies utilized the dermal or skin and the pulmonary exposures as the primary routes of toxicity. It was interesting that only very few studies revealed that the TiO2 nanoparticles could penetrate through the skin and translocate to other tissues, while many other studies demonstrated that no penetration or translocation could happen through the skin. Conversely, the TiO2 nanoparticles that entered through the pulmonary route were translocated to the brain or the systemic circulation from where these reached other organs like the kidney, liver, etc. In most studies, TiO2 nanoparticles appeared to have caused oxidative stress, histopathological alterations, carcinogenesis, genotoxicity and immune disruption. Therefore, the use of such materials in humans must be either avoided or strictly managed to minimise risks for human health in various situations.
Collapse
Affiliation(s)
- Muhammad Shakeel
- Department of Zoology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Farhat Jabeen
- Department of Zoology, Government College University Faisalabad, Faisalabad, Pakistan.
| | - Samina Shabbir
- Department of Zoology, Government College University Faisalabad, Faisalabad, Pakistan
| | | | - Muhammad Saleem Khan
- Department of Zoology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Abdul Shakoor Chaudhry
- School of Agriculture, Food and Rural Development, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| |
Collapse
|
33
|
Yu X, Hong F, Zhang YQ. Bio-effect of nanoparticles in the cardiovascular system. J Biomed Mater Res A 2016; 104:2881-97. [PMID: 27301683 DOI: 10.1002/jbm.a.35804] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/07/2016] [Indexed: 12/21/2022]
Abstract
Nanoparticles (NPs; < 100 nm) are increasingly being applied in various fields due to their unique physicochemical properties. The increase in human exposure to NPs has raised concerns regarding their health and safety profiles. The potential correlation between NP exposure and several cardiovascular (CV) events has been demonstrated. The aim of this review is to provide a comprehensive evaluation of the current knowledge regarding the bio-toxic impacts of titanium oxide, silver, silica, carbon black, carbon nanotube, and zinc oxide NPs exposure on the CV system in terms of in vivo and in vitro experiments, which is not fully understood presently. Moreover, the potential toxic mechanisms of NPs in the CV system that are still being questioned are elaborately discussed, and the underlying capacity of NPs used in medicine for CV events are summarized. It will be an important instrument to extrapolate relevant data for human CV risk evaluation and management. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 2881-2897, 2016.
Collapse
Affiliation(s)
- Xiaohong Yu
- Department of Applied Biology, School of Basic Medical and Biological Sciences, Soochow University, RM702-2303, Renai Road No. 199, Dushuhu Higher Edu. Town, Suzhou, 215123, People's Republic of China
| | - Fashui Hong
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian, 223300, China. .,Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian, 223300, China.
| | - Yu-Qing Zhang
- Department of Applied Biology, School of Basic Medical and Biological Sciences, Soochow University, RM702-2303, Renai Road No. 199, Dushuhu Higher Edu. Town, Suzhou, 215123, People's Republic of China
| |
Collapse
|
34
|
Aragon M, Erdely A, Bishop L, Salmen R, Weaver J, Liu J, Hall P, Eye T, Kodali V, Zeidler-Erdely P, Stafflinger JE, Ottens AK, Campen MJ. MMP-9-Dependent Serum-Borne Bioactivity Caused by Multiwalled Carbon Nanotube Exposure Induces Vascular Dysfunction via the CD36 Scavenger Receptor. Toxicol Sci 2016; 150:488-98. [PMID: 26801584 PMCID: PMC4966280 DOI: 10.1093/toxsci/kfw015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Inhalation of multiwalled carbon nanotubes (MWCNT) causes systemic effects including vascular inflammation, endothelial dysfunction, and acute phase protein expression. MWCNTs translocate only minimally beyond the lungs, thus cardiovascular effects thereof may be caused by generation of secondary biomolecular factors from MWCNT-pulmonary interactions that spill over into the systemic circulation. Therefore, we hypothesized that induced matrix metalloproteinase-9 (MMP-9) is a generator of factors that, in turn, drive vascular effects through ligand-receptor interactions with the multiligand pattern recognition receptor, CD36. To test this, wildtype (WT; C57BL/6) and MMP-9(-/-)mice were exposed to varying doses (10 or 40 µg) of MWCNTs via oropharyngeal aspiration and serum was collected at 4 and 24 h postexposure. Endothelial cells treated with serum from MWCNT-exposed WT mice exhibited significantly reduced nitric oxide (NO) generation, as measured by electron paramagnetic resonance, an effect that was independent of NO scavenging. Serum from MWCNT-exposed WT mice inhibited acetylcholine (ACh)-mediated relaxation of aortic rings at both time points. Absence of CD36 on the aortic rings (obtained from CD36-deficient mice) abolished the serum-induced impairment of vasorelaxation. MWCNT exposure induced MMP-9 protein levels in both bronchoalveolar lavage and whole lung lysates. Serum from MMP-9(-/-)mice exposed to MWCNT did not diminish the magnitude of vasorelaxation in naïve WT aortic rings, although a modest right shift of the ACh dose-response curve was observed in both MWCNT dose groups relative to controls. In conclusion, pulmonary exposure to MWCNT leads to elevated MMP-9 levels and MMP-9-dependent generation of circulating bioactive factors that promote endothelial dysfunction and decreased NO bioavailability via interaction with vascular CD36.
Collapse
Affiliation(s)
- Mario Aragon
- *Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, New Mexico 87131
| | - Aaron Erdely
- National Institute for Occupational Safety and Health, Morgantown, West Virginia 26508
| | - Lindsey Bishop
- National Institute for Occupational Safety and Health, Morgantown, West Virginia 26508
| | - Rebecca Salmen
- National Institute for Occupational Safety and Health, Morgantown, West Virginia 26508
| | - John Weaver
- *Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, New Mexico 87131
| | - Jim Liu
- *Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, New Mexico 87131
| | - Pamela Hall
- *Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, New Mexico 87131
| | - Tracy Eye
- National Institute for Occupational Safety and Health, Morgantown, West Virginia 26508
| | - Vamsi Kodali
- National Institute for Occupational Safety and Health, Morgantown, West Virginia 26508
| | - Patti Zeidler-Erdely
- National Institute for Occupational Safety and Health, Morgantown, West Virginia 26508
| | - Jillian E Stafflinger
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298
| | - Andrew K Ottens
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298
| | - Matthew J Campen
- *Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, New Mexico 87131
| |
Collapse
|
35
|
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]
|
36
|
Clark J, Gregory CC, Matthews IP, Hoogendoorn B. The biological effects upon the cardiovascular system consequent to exposure to particulates of less than 500 nm in size. Biomarkers 2015; 21:1-47. [PMID: 26643755 DOI: 10.3109/1354750x.2015.1118540] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT Ultrafine particulate matter contribution to cardiovascular disease is not known and not regulated. PM up to 500 nm are abundant in urban air and alveolar deposition is significant. OBJECTIVE Effects beyond the alveolar barrier within the body or in vitro tissues exposed to particles <500 nm. METHODS AND RESULTS DATABASES MEDLINE; Ovid-MEDLINE PREM; Web of Science; PubMed (SciGlobe). 127 articles. Results in tables: "subject type exposed", "exposure type", "technique". CONCLUSION Heart rate, vasoactivity, atherosclerotic advancement, oxidative stress, coagulability, inflammatory changes are affected. Production of reactive oxygen species is a useful target to limit outcomes associated with UFP exposure.
Collapse
Affiliation(s)
- James Clark
- a Institute of Primary Care and Public Health, School of Medicine, Cardiff University , Cardiff , UK
| | - Clive C Gregory
- a Institute of Primary Care and Public Health, School of Medicine, Cardiff University , Cardiff , UK
| | - Ian P Matthews
- a Institute of Primary Care and Public Health, School of Medicine, Cardiff University , Cardiff , UK
| | - Bastiaan Hoogendoorn
- a Institute of Primary Care and Public Health, School of Medicine, Cardiff University , Cardiff , UK
| |
Collapse
|
37
|
Wauters A, Vicenzi M, De Becker B, Riga JP, Esmaeilzadeh F, Faoro V, Vachiéry JL, van de Borne P, Argacha JF. At high cardiac output, diesel exhaust exposure increases pulmonary vascular resistance and decreases distensibility of pulmonary resistive vessels. Am J Physiol Heart Circ Physiol 2015; 309:H2137-44. [PMID: 26497960 DOI: 10.1152/ajpheart.00149.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 10/19/2015] [Indexed: 12/19/2022]
Abstract
Air pollution has recently been associated with the development of acute decompensated heart failure, but the underlying biological mechanisms remain unclear. A pulmonary vasoconstrictor effect of air pollution, combined with its systemic effects, may precipitate decompensated heart failure. The aim of the present study was to investigate the effects of acute exposure to diesel exhaust (DE) on pulmonary vascular resistance (PVR) under resting and stress conditions but also to determine whether air pollution may potentiate acquired pulmonary hypertension. Eighteen healthy male volunteers were exposed to ambient air (AA) or dilute DE with a particulate matter of <2.5 μm concentration of 300 μg/m(3) for 2 h in a randomized, crossover study design. The effects of DE on PVR, on the coefficient of distensibilty of pulmonary vessels (α), and on right and left ventricular function were evaluated at rest (n = 18), during dobutamine stress echocardiography (n = 10), and during exercise stress echocardiography performed in hypoxia (n = 8). Serum endothelin-1 and fractional exhaled nitric oxide were also measured. At rest, exposure to DE did not affect PVR. During dobutamine stress, the slope of the mean pulmonary artery pressure-cardiac output relationship increased from 2.8 ± 0.5 mmHg · min · l (-1) in AA to 3.9 ± 0.5 mmHg · min · l (-1) in DE (P < 0.05) and the α coefficient decreased from 0.96 ± 0.15 to 0.64 ± 0.12%/mmHg (P < 0.01). DE did not further enhance the hypoxia-related upper shift of the mean pulmonary artery pressure-cardiac output relationship. Exposure to DE did not affect serum endothelin-1 concentration or fractional exhaled nitric oxide. In conclusion, acute exposure to DE increased pulmonary vasomotor tone by decreasing the distensibility of pulmonary resistive vessels at high cardiac output.
Collapse
Affiliation(s)
- Aurélien Wauters
- Department of Cardiology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium;
| | - Marco Vicenzi
- Department of Cardiology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium; Laboratory of Physiology and Physiopathology, Université Libre de Bruxellesm, Brussels, Belgium; and
| | - Benjamin De Becker
- Department of Cardiology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Philippe Riga
- Department of Cardiology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Fatemeh Esmaeilzadeh
- Department of Cardiology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Vitalie Faoro
- Laboratory of Physiology and Physiopathology, Université Libre de Bruxellesm, Brussels, Belgium; and
| | - Jean-Luc Vachiéry
- Department of Cardiology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Philippe van de Borne
- Department of Cardiology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-François Argacha
- Department of Cardiology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium; Department of Cardiology, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
38
|
Stapleton PA, McBride CR, Yi J, Nurkiewicz TR. Uterine microvascular sensitivity to nanomaterial inhalation: An in vivo assessment. Toxicol Appl Pharmacol 2015; 288:420-8. [PMID: 26375943 DOI: 10.1016/j.taap.2015.08.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 07/30/2015] [Accepted: 08/19/2015] [Indexed: 12/21/2022]
Abstract
With the tremendous number and diverse applications of engineered nanomaterials incorporated in daily human activity, exposure can no longer be solely confined to occupational exposures of healthy male models. Cardiovascular and endothelial cell dysfunction have been established using in vitro and in situ preparations, but the translation to intact in vivo models is limited. Intravital microscopy has been used extensively to understand microvascular physiology while maintaining in vivo neurogenic, humoral, and myogenic control. However, a tissue specific model to assess the influences of nanomaterial exposure on female reproductive health has not been fully elucidated. Female Sprague Dawley (SD) rats were exposed to nano-TiO2 aerosols (171 ± 6 nm, 10.1 ± 0.39 mg/m(3), 5h) 24-hours prior to experimentation, leading to a calculated deposition of 42.0 ± 1.65 μg. After verifying estrus status, vital signs were monitored and the right horn of the uterus was exteriorized, gently secured over an optical pedestal, and enclosed in a warmed tissue bath using intravital microscopy techniques. After equilibration, significantly higher leukocyte-endothelium interactions were recorded in the exposed group. Arteriolar responsiveness was assessed using ionophoretically applied agents: muscarinic agonist acetylcholine (0.025 M; ACh; 20, 40, 100, and 200 nA), and nitric oxide donor sodium nitroprusside (0.05 M; SNP; 20, 40, and 100 nA), or adrenergic agonist phenylephrine (0.05 M; PE; 20, 40, and 100 nA) using glass micropipettes. Passive diameter was established by tissue superfusion with 10(-4)M adenosine. Similar to male counterparts, female SD rats present systemic microvascular dysfunction; however the ramifications associated with female health and reproduction have yet to be elucidated.
Collapse
Affiliation(s)
- P A Stapleton
- Center for Cardiovascular and Respiratory Sciences, West Virginia University School of Medicine, Morgantown, WV 26506, United States; Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV 26506, United States
| | - C R McBride
- Center for Cardiovascular and Respiratory Sciences, West Virginia University School of Medicine, Morgantown, WV 26506, United States; Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV 26506, United States
| | - J Yi
- Center for Cardiovascular and Respiratory Sciences, West Virginia University School of Medicine, Morgantown, WV 26506, United States; Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV 26506, United States
| | - T R Nurkiewicz
- Center for Cardiovascular and Respiratory Sciences, West Virginia University School of Medicine, Morgantown, WV 26506, United States; Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV 26506, United States.
| |
Collapse
|
39
|
Maurer MM, Donohoe GC, Maleki H, Yi J, McBride C, Nurkiewicz TR, Valentine SJ. Comparative plasma proteomic studies of pulmonary TiO2 nanoparticle exposure in rats using liquid chromatography tandem mass spectrometry. J Proteomics 2015; 130:85-93. [PMID: 26375203 DOI: 10.1016/j.jprot.2015.09.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 08/14/2015] [Accepted: 09/05/2015] [Indexed: 11/17/2022]
Abstract
Mounting evidence suggests that pulmonary exposure to nanoparticles (NPs) has a toxic effect on biological systems. A number of studies have shown that exposure to NPs result in systemic inflammatory response, oxidative stress, and leukocyte adhesion. However, significant knowledge gaps exist for understanding the key molecular mechanisms responsible for altered microvasculature function. Utilizing comprehensive LC-MS/MS and comparative proteomic analysis strategies, important proteins related to TiO2 NP exposure in rat plasma have been identified. Molecular pathway analysis of these proteins revealed 13 canonical pathways as being significant (p ≤ 0.05), but none were found to be significantly up or down-regulated (z>|2|). This work lays the foundation for future research that will monitor relative changes in protein abundance in plasma and tissue as a function of post-exposure time and TiO2 NP dosage to further elucidate mechanisms of pathway activation as well as to decipher other affected pathways.
Collapse
Affiliation(s)
- Megan M Maurer
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV 26506, United States
| | - Gregory C Donohoe
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV 26506, United States
| | - Hossein Maleki
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV 26506, United States
| | - Jinghai Yi
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, United States
| | - Carroll McBride
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, United States
| | - Timothy R Nurkiewicz
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, United States
| | - Stephen J Valentine
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV 26506, United States.
| |
Collapse
|
40
|
Stapleton PA, Nichols CE, Yi J, McBride CR, Minarchick VC, Shepherd DL, Hollander JM, Nurkiewicz TR. Microvascular and mitochondrial dysfunction in the female F1 generation after gestational TiO2 nanoparticle exposure. Nanotoxicology 2015; 9:941-51. [PMID: 25475392 DOI: 10.3109/17435390.2014.984251] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Due to the ongoing evolution of nanotechnology, there is a growing need to assess the toxicological outcomes in under-studied populations in order to properly consider the potential of engineered nanomaterials (ENM) and fully enhance their safety. Recently, we and others have explored the vascular consequences associated with gestational nanomaterial exposure, reporting microvascular dysfunction within the uterine circulation of pregnant dams and the tail artery of fetal pups. It has been proposed (via work derived by the Barker Hypothesis) that mitochondrial dysfunction and subsequent oxidative stress mechanisms as a possible link between a hostile gestational environment and adult disease. Therefore, in this study, we exposed pregnant Sprague-Dawley rats to nanosized titanium dioxide aerosols after implantation (gestational day 6). Pups were delivered, and the progeny grew into adulthood. Microvascular reactivity, mitochondrial respiration and hydrogen peroxide production of the coronary and uterine circulations of the female offspring were evaluated. While there were no significant differences within the maternal or litter characteristics, endothelium-dependent dilation and active mechanotransduction in both coronary and uterine arterioles were significantly impaired. In addition, there was a significant reduction in maximal mitochondrial respiration (state 3) in the left ventricle and uterus. These studies demonstrate microvascular dysfunction and coincide with mitochondrial inefficiencies in both the cardiac and uterine tissues, which may represent initial evidence that prenatal ENM exposure produces microvascular impairments that persist throughout multiple developmental stages.
Collapse
Affiliation(s)
- Phoebe A Stapleton
- a Center for Cardiovascular and Respiratory Sciences .,b Department of Physiology and Pharmacology , and
| | - Cody E Nichols
- a Center for Cardiovascular and Respiratory Sciences .,c Division of Exercise Physiology , West Virginia University School of Medicine , Morgantown , WV , USA
| | - Jinghai Yi
- a Center for Cardiovascular and Respiratory Sciences .,b Department of Physiology and Pharmacology , and
| | - Carroll R McBride
- a Center for Cardiovascular and Respiratory Sciences .,b Department of Physiology and Pharmacology , and
| | - Valerie C Minarchick
- a Center for Cardiovascular and Respiratory Sciences .,b Department of Physiology and Pharmacology , and
| | - Danielle L Shepherd
- a Center for Cardiovascular and Respiratory Sciences .,c Division of Exercise Physiology , West Virginia University School of Medicine , Morgantown , WV , USA
| | - John M Hollander
- a Center for Cardiovascular and Respiratory Sciences .,c Division of Exercise Physiology , West Virginia University School of Medicine , Morgantown , WV , USA
| | - Timothy R Nurkiewicz
- a Center for Cardiovascular and Respiratory Sciences .,b Department of Physiology and Pharmacology , and
| |
Collapse
|
41
|
Sha B, Gao W, Cui X, Wang L, Xu F. The potential health challenges of TiO2nanomaterials. J Appl Toxicol 2015; 35:1086-101. [DOI: 10.1002/jat.3193] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 05/10/2015] [Accepted: 05/10/2015] [Indexed: 01/09/2023]
Affiliation(s)
- Baoyong Sha
- School of Basic Medical Science; Xi'an Medical University; Xi'an 710021 China
- Bioinspired Engineering & Biomechanics Center (BEBC); Xi'an Jiaotong University; Xi'an 710049 China
| | - Wei Gao
- Department of Anesthesiology; the First Affiliated Hospital of Xi'an Jiaotong University Health Science Center; Xi'an 710061 China
| | - Xingye Cui
- Bioinspired Engineering & Biomechanics Center (BEBC); Xi'an Jiaotong University; Xi'an 710049 China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology; Xi'an Jiaotong University; Xi'an 710049 China
| | - Lin Wang
- Bioinspired Engineering & Biomechanics Center (BEBC); Xi'an Jiaotong University; Xi'an 710049 China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology; Xi'an Jiaotong University; Xi'an 710049 China
| | - Feng Xu
- Bioinspired Engineering & Biomechanics Center (BEBC); Xi'an Jiaotong University; Xi'an 710049 China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology; Xi'an Jiaotong University; Xi'an 710049 China
| |
Collapse
|
42
|
Li H, Hedmer M, Kåredal M, Björk J, Stockfelt L, Tinnerberg H, Albin M, Broberg K. A Cross-Sectional Study of the Cardiovascular Effects of Welding Fumes. PLoS One 2015; 10:e0131648. [PMID: 26147298 PMCID: PMC4492943 DOI: 10.1371/journal.pone.0131648] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 06/02/2015] [Indexed: 12/13/2022] Open
Abstract
Objectives Occupational exposure to particulate air pollution has been associated with an increased risk of cardiovascular disease. However, the risk to welders working today remains unclear. We aimed to elucidate the cardiovascular effects of exposure to welding fumes. Methods In a cross-sectional study, structured interviews and biological sampling were conducted for 101 welders and 127 controls (all non-smoking males) from southern Sweden. Personal breathing zone sampling of respirable dust was performed. Blood pressure (BP) and endothelial function (using peripheral arterial tonometry) were measured. Plasma and serum samples were collected from peripheral blood for measurement of C-reactive protein, low-density lipoprotein, homocysteine, serum amyloid A, and cytokines. Results Welders were exposed to 10-fold higher levels of particles than controls. Welders had significantly higher BP compared to controls, an average of 5 mm Hg higher systolic and diastolic BP (P≤0.001). IL-8 was 3.4 ng/L higher in welders (P=0.010). Years working as a welder were significantly associated with increased BP (β=0.35, 95%CI 0.13 – 0.58, P=0.0024 for systolic BP; β=0.32, 95%CI 0.16 – 0.48, P<0.001 for diastolic BP, adjusted for BMI) but exposure to respirable dust was not associated with BP. No clear associations occurred between welding and endothelial function, or other effect markers. Conclusions A modest increase in BP was found among welders compared to controls suggesting that low-to-moderate exposure to welding fumes remains a risk factor for cardiovascular disease.
Collapse
Affiliation(s)
- Huiqi Li
- Division of Occupational and Environmental Medicine, Laboratory Medicine, Lund University, Lund, Sweden
| | - Maria Hedmer
- Division of Occupational and Environmental Medicine, Laboratory Medicine, Lund University, Lund, Sweden
| | - Monica Kåredal
- Division of Occupational and Environmental Medicine, Laboratory Medicine, Lund University, Lund, Sweden
| | - Jonas Björk
- Competence Centre for Clinical Research, Lund University, Lund, Sweden
| | - Leo Stockfelt
- Department of Occupational and Environmental Medicine, Sahlgrenska University Hospital and Academy, University of Gothenburg, Gothenburg, Sweden
| | - Håkan Tinnerberg
- Division of Occupational and Environmental Medicine, Laboratory Medicine, Lund University, Lund, Sweden
| | - Maria Albin
- Division of Occupational and Environmental Medicine, Laboratory Medicine, Lund University, Lund, Sweden
| | - Karin Broberg
- Division of Occupational and Environmental Medicine, Laboratory Medicine, Lund University, Lund, Sweden
- Unit of Metals & Health, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
43
|
Paffett ML, Zychowski KE, Sheppard L, Robertson S, Weaver JM, Lucas SN, Campen MJ. Ozone Inhalation Impairs Coronary Artery Dilation via Intracellular Oxidative Stress: Evidence for Serum-Borne Factors as Drivers of Systemic Toxicity. Toxicol Sci 2015; 146:244-53. [PMID: 25962394 DOI: 10.1093/toxsci/kfv093] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ambient ozone (O3) levels are associated with cardiovascular morbidity and mortality, but the underlying pathophysiological mechanisms driving extrapulmonary toxicity remain unclear. This study examined the coronary vascular bed of rats in terms of constrictive and dilatory responses to known agonists following a single O3 inhalation exposure. In addition, serum from exposed rats was used in ex vivo preparations to examine whether bioactivity and toxic effects of inhaled O3 could be conveyed to extrapulmonary systems via the circulation. We found that 24 h following inhalation of 1 ppm O3, isolated coronary vessels exhibited greater basal tone and constricted to a greater degree to serotonin stimulation. Vasodilation to acetylcholine (ACh) was markedly diminished in coronary arteries from O3-exposed rats, compared with filtered air-exposed controls. Dilation to ACh was restored by combined superoxide dismutase and catalase treatment, and also by NADPH oxidase inhibition. When dilute (10%) serum from exposed rats was perfused into the lumen of coronary arteries from unexposed, naïve rats, the O3-induced reduction in vasodilatory response to ACh was partially recapitulated. Furthermore, following O3 inhalation, serum exhibited a nitric oxide scavenging capacity, which may partially explain blunted ACh-mediated vasodilatory responses. Thus, bioactivity from inhalation exposures may be due to compositional changes of the circulation. These studies shed light on possible mechanisms of action that may explain O3-associated cardiac morbidity and mortality in humans.
Collapse
Affiliation(s)
- Michael L Paffett
- *Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico
| | - Katherine E Zychowski
- *Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico
| | - Lianne Sheppard
- Departments of Biostatistics and Environmental & Occupational Health Sciences, School of Public Health, University of Washington, Seattle, Washington and
| | - Sarah Robertson
- Toxicology Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Chilton, UK
| | - John M Weaver
- *Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico
| | - Selita N Lucas
- *Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico
| | - Matthew J Campen
- *Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico,
| |
Collapse
|
44
|
Armstead AL, Minarchick VC, Porter DW, Nurkiewicz TR, Li B. Acute inflammatory responses of nanoparticles in an intra-tracheal instillation rat model. PLoS One 2015; 10:e0118778. [PMID: 25738830 PMCID: PMC4349695 DOI: 10.1371/journal.pone.0118778] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 01/22/2015] [Indexed: 12/30/2022] Open
Abstract
Exposure to hard metal tungsten carbide cobalt (WC-Co) "dusts" in enclosed industrial environments is known to contribute to the development of hard metal lung disease and an increased risk for lung cancer. Currently, the influence of local and systemic inflammation on disease progression following WC-Co exposure remains unclear. To better understand the relationship between WC-Co nanoparticle (NP) exposure and its resultant effects, the acute local pulmonary and systemic inflammatory responses caused by WC-Co NPs were explored using an intra-tracheal instillation (IT) model and compared to those of CeO2 (another occupational hazard) NP exposure. Sprague-Dawley rats were given an IT dose (0-500 μg per rat) of WC-Co or CeO2 NPs. Following 24-hr exposure, broncho-alveolar lavage fluid and whole blood were collected and analyzed. A consistent lack of acute local pulmonary inflammation was observed in terms of the broncho-alveolar lavage fluid parameters examined (i.e. LDH, albumin, and macrophage activation) in animals exposed to WC-Co NP; however, significant acute pulmonary inflammation was observed in the CeO2 NP group. The lack of acute inflammation following WC-Co NP exposure contrasts with earlier in vivo reports regarding WC-Co toxicity in rats, illuminating the critical role of NP dose and exposure time and bringing into question the potential role of impurities in particle samples. Further, we demonstrated that WC-Co NP exposure does not induce acute systemic effects since no significant increase in circulating inflammatory cytokines were observed. Taken together, the results of this in vivo study illustrate the distinct differences in acute local pulmonary and systemic inflammatory responses to NPs composed of WC-Co and CeO2; therefore, it is important that the outcomes of pulmonary exposure to one type of NPs may not be implicitly extrapolated to other types of NPs.
Collapse
Affiliation(s)
- Andrea L. Armstead
- Biomaterials, Bioengineering & Nanotechnology Laboratory, Department of Orthopaedics, School of Medicine, West Virginia University, Morgantown, West Virginia, United States of America
- Pharmaceutical and Pharmacological Sciences Graduate Program, School of Pharmacy, West Virginia University, Morgantown, West Virginia, United States of America
| | - Valerie C. Minarchick
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, Morgantown, West Virginia, United States of America
- Center for Cardiovascular and Respiratory Sciences, Robert C. Byrd Health Sciences Center, School of Medicine, West Virginia University, Morgantown, West Virginia, United States of America
| | - Dale W. Porter
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, Morgantown, West Virginia, United States of America
- Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia, United States of America
| | - Timothy R. Nurkiewicz
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, Morgantown, West Virginia, United States of America
- Center for Cardiovascular and Respiratory Sciences, Robert C. Byrd Health Sciences Center, School of Medicine, West Virginia University, Morgantown, West Virginia, United States of America
- Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia, United States of America
| | - Bingyun Li
- Biomaterials, Bioengineering & Nanotechnology Laboratory, Department of Orthopaedics, School of Medicine, West Virginia University, Morgantown, West Virginia, United States of America
- Pharmaceutical and Pharmacological Sciences Graduate Program, School of Pharmacy, West Virginia University, Morgantown, West Virginia, United States of America
- Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia, United States of America
- Mary Babb Randolph Cancer Center, Morgantown, West Virginia, United States of America
- * E-mail:
| |
Collapse
|
45
|
Haberl N, Hirn S, Holzer M, Zuchtriegel G, Rehberg M, Krombach F. Effects of acute systemic administration of TiO2, ZnO, SiO2, and Ag nanoparticles on hemodynamics, hemostasis and leukocyte recruitment. Nanotoxicology 2015; 9:963-71. [DOI: 10.3109/17435390.2014.992815] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
46
|
Oberdörster G, Castranova V, Asgharian B, Sayre P. Inhalation Exposure to Carbon Nanotubes (CNT) and Carbon Nanofibers (CNF): Methodology and Dosimetry. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2015; 18:121-212. [PMID: 26361791 PMCID: PMC4706753 DOI: 10.1080/10937404.2015.1051611] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Carbon nanotubes (CNT) and nanofibers (CNF) are used increasingly in a broad array of commercial products. Given current understandings, the most significant life-cycle exposures to CNT/CNF occur from inhalation when they become airborne at different stages of their life cycle, including workplace, use, and disposal. Increasing awareness of the importance of physicochemical properties as determinants of toxicity of CNT/CNF and existing difficulties in interpreting results of mostly acute rodent inhalation studies to date necessitate a reexamination of standardized inhalation testing guidelines. The current literature on pulmonary exposure to CNT/CNF and associated effects is summarized; recommendations and conclusions are provided that address test guideline modifications for rodent inhalation studies that will improve dosimetric extrapolation modeling for hazard and risk characterization based on the analysis of exposure-dose-response relationships. Several physicochemical parameters for CNT/CNF, including shape, state of agglomeration/aggregation, surface properties, impurities, and density, influence toxicity. This requires an evaluation of the correlation between structure and pulmonary responses. Inhalation, using whole-body exposures of rodents, is recommended for acute to chronic pulmonary exposure studies. Dry powder generator methods for producing CNT/CNF aerosols are preferred, and specific instrumentation to measure mass, particle size and number distribution, and morphology in the exposure chambers are identified. Methods are discussed for establishing experimental exposure concentrations that correlate with realistic human exposures, such that unrealistically high experimental concentrations need to be identified that induce effects under mechanisms that are not relevant for workplace exposures. Recommendations for anchoring data to results seen for positive and negative benchmark materials are included, as well as periods for postexposure observation. A minimum data set of specific bronchoalveolar lavage parameters is recommended. Retained lung burden data need to be gathered such that exposure-dose-response correlations may be analyzed and potency comparisons between materials and mammalian species are obtained considering dose metric parameters for interpretation of results. Finally, a list of research needs is presented to fill data gaps for further improving design, analysis, and interpretation and extrapolation of results of rodent inhalation studies to refine meaningful risk assessments for humans.
Collapse
Affiliation(s)
- Günter Oberdörster
- Department of Environmental Medicine, University of Rochester, Rochester, New York, USA
| | - Vincent Castranova
- Formerly with the National Institute for Occupational Safety and Health, West Virginia University School of Pharmacy, Morgantown, West Virginia, USA
| | | | - Phil Sayre
- Formerly with the U.S. Environmental Protection Agency, Washington, DC, USA
| |
Collapse
|
47
|
Weichenthal S, Hatzopoulou M, Goldberg MS. Exposure to traffic-related air pollution during physical activity and acute changes in blood pressure, autonomic and micro-vascular function in women: a cross-over study. Part Fibre Toxicol 2014; 11:70. [PMID: 25487431 PMCID: PMC4276095 DOI: 10.1186/s12989-014-0070-4] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 11/24/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Traffic-related air pollution may contribute to cardiovascular morbidity. In urban areas, exposures during physical activity are of interest owing to increased breathing rates and close proximity to vehicle emissions. METHODS We conducted a cross-over study among 53 healthy non-smoking women in Montreal, Canada during the summer of 2013. Women were exposed to traffic pollutants for 2-hours on three separate occasions during cycling on high and low-traffic routes as well as indoors. Personal air pollution exposures (PM(2.5), ultrafine particles (UFP), black carbon, NO₂, and O₃) were evaluated along each route and linear mixed-effects models with random subject intercepts were used to estimate the impact of air pollutants on acute changes in blood pressure, heart rate variability, and micro-vascular function in the hours immediately following exposure. Single and multi-pollutant models were examined and potential effect modification by mean regional air pollution concentrations (PM(2.5), NO₂, and O₃) was explored for the 24-hour and 5-day periods preceding exposure. RESULTS In total, 143 exposure routes were completed. Each interquartile increase (10,850/cm³) in UFP exposure was associated with a 4.91% (95% CI: -9.31, -0.512) decrease in reactive hyperemia index (a measure of micro-vascular function) and each 24 ppb increase in O₃ exposure corresponded to a 2.49% (95% CI: 0.141, 4.84) increase in systolic blood pressure and a 3.26% (95% CI: 0.0117, 6.51) increase in diastolic blood pressure 3-hours after exposure. Personal exposure to PM(2.5) was associated with decreases in HRV measures reflecting parasympathetic modulation of the heart and regional PM(2.5) concentrations modified these relationships (p < 0.05). In particular, stronger inverse associations were observed when regional PM(2.5) was higher on the days prior to the study period. Regional PM(2.5) also modified the impact of personal O₃ on the standard deviation of normal to normal intervals (SDNN) (p < 0.05): a significant inverse relationship was observed when regional PM(2.5) was low prior to study periods and a significant positive relationship was observed when regional PM(2.5) was high. CONCLUSION Exposure to traffic pollution may contribute to acute changes in blood pressure, autonomic and micro-vascular function in women. Regional air pollution concentrations may modify the impact of these exposures on autonomic function.
Collapse
Affiliation(s)
- Scott Weichenthal
- Air Health Science Division, Health Canada, 269 Laurier Avenue West, K1A 0K9, Ottawa, ON, Canada.
| | - Marianne Hatzopoulou
- Department of Civil Engineering, McGill University, Macdonald Engineering Building, 817 Sherbrooke Street West, H3A 0C3, Montreal, Quebec, Canada.
| | - Mark S Goldberg
- Division of Clinical Epidemiology, McGill University Health Center, 687 Pine Avenue West, H3A 1A1, Montreal, Quebec, Canada.
| |
Collapse
|
48
|
Minarchick VC, Stapleton PA, Fix NR, Leonard SS, Sabolsky EM, Nurkiewicz TR. Intravenous and gastric cerium dioxide nanoparticle exposure disrupts microvascular smooth muscle signaling. Toxicol Sci 2014; 144:77-89. [PMID: 25481005 DOI: 10.1093/toxsci/kfu256] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cerium dioxide nanoparticles (CeO2 NP) hold great therapeutic potential, but the in vivo effects of non-pulmonary exposure routes are unclear. The first aim was to determine whether microvascular function is impaired after intravenous and gastric CeO2 NP exposure. The second aim was to investigate the mechanism(s) of action underlying microvascular dysfunction following CeO2 NP exposure. Rats were exposed to CeO2 NP (primary diameter: 4 ± 1 nm, surface area: 81.36 m(2)/g) by intratracheal instillation, intravenous injection, or gastric gavage. Mesenteric arterioles were harvested 24 h post-exposure and vascular function was assessed using an isolated arteriole preparation. Endothelium-dependent and independent function and vascular smooth muscle (VSM) signaling (soluble guanylyl cyclase [sGC] and cyclic guanosine monophosphate [cGMP]) were assessed. Reactive oxygen species (ROS) generation and nitric oxide (NO) production were analyzed. Compared with controls, endothelium-dependent and independent dilation were impaired following intravenous injection (by 61% and 45%) and gastric gavage (by 63% and 49%). However, intravenous injection resulted in greater microvascular impairment (16% and 35%) compared with gastric gavage at an identical dose (100 µg). Furthermore, sGC activation and cGMP responsiveness were impaired following pulmonary, intravenous, and gastric CeO2 NP treatment. Finally, nanoparticle exposure resulted in route-dependent, increased ROS generation and decreased NO production. These results indicate that CeO2 NP exposure route differentially impairs microvascular function, which may be mechanistically linked to decreased NO production and subsequent VSM signaling. Fully understanding the mechanisms behind CeO2 NP in vivo effects is a critical step in the continued therapeutic development of this nanoparticle.
Collapse
Affiliation(s)
- Valerie C Minarchick
- *Center for Cardiovascular and Respiratory Sciences and Department of Physiology and Pharmacology, West Virginia University School of Medicine, Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health and Department of Mechanical and Aerospace Engineering, West Virginia University, Morgantown, West Virginia 26506 *Center for Cardiovascular and Respiratory Sciences and Department of Physiology and Pharmacology, West Virginia University School of Medicine, Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health and Department of Mechanical and Aerospace Engineering, West Virginia University, Morgantown, West Virginia 26506
| | - Phoebe A Stapleton
- *Center for Cardiovascular and Respiratory Sciences and Department of Physiology and Pharmacology, West Virginia University School of Medicine, Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health and Department of Mechanical and Aerospace Engineering, West Virginia University, Morgantown, West Virginia 26506 *Center for Cardiovascular and Respiratory Sciences and Department of Physiology and Pharmacology, West Virginia University School of Medicine, Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health and Department of Mechanical and Aerospace Engineering, West Virginia University, Morgantown, West Virginia 26506
| | - Natalie R Fix
- *Center for Cardiovascular and Respiratory Sciences and Department of Physiology and Pharmacology, West Virginia University School of Medicine, Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health and Department of Mechanical and Aerospace Engineering, West Virginia University, Morgantown, West Virginia 26506
| | - Stephen S Leonard
- *Center for Cardiovascular and Respiratory Sciences and Department of Physiology and Pharmacology, West Virginia University School of Medicine, Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health and Department of Mechanical and Aerospace Engineering, West Virginia University, Morgantown, West Virginia 26506
| | - Edward M Sabolsky
- *Center for Cardiovascular and Respiratory Sciences and Department of Physiology and Pharmacology, West Virginia University School of Medicine, Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health and Department of Mechanical and Aerospace Engineering, West Virginia University, Morgantown, West Virginia 26506
| | - Timothy R Nurkiewicz
- *Center for Cardiovascular and Respiratory Sciences and Department of Physiology and Pharmacology, West Virginia University School of Medicine, Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health and Department of Mechanical and Aerospace Engineering, West Virginia University, Morgantown, West Virginia 26506 *Center for Cardiovascular and Respiratory Sciences and Department of Physiology and Pharmacology, West Virginia University School of Medicine, Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health and Department of Mechanical and Aerospace Engineering, West Virginia University, Morgantown, West Virginia 26506 *Center for Cardiovascular and Respiratory Sciences and Department of Physiology and Pharmacology, West Virginia University School of Medicine, Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health and Department of Mechanical and Aerospace Engineering, West Virginia University, Morgantown, West Virginia 26506
| |
Collapse
|
49
|
Huang KT, Wu CT, Huang KH, Lin WC, Chen CM, Guan SS, Chiang CK, Liu SH. Titanium nanoparticle inhalation induces renal fibrosis in mice via an oxidative stress upregulated transforming growth factor-β pathway. Chem Res Toxicol 2014; 28:354-64. [PMID: 25406100 DOI: 10.1021/tx500287f] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Titanium dioxide nanoparticles (Nano-TiO2) are gradually being used extensively in clinical settings, industry, and daily life. Accumulation studies showed that Nano-TiO2 exposure is able to cause injuries in various animal organs, including the lung, liver, spleen, and kidney. However, it remains unclear whether exposure of Nano-TiO2 by inhalation causes renal fibrosis. Here, we investigated the role of reactive oxygen species (ROS)/reactive nitrogen species (RNS) related signaling molecules in chronic renal damage after Nano-TiO2 inhalation in mice. Mice were treated with Nano-TiO2 (0.1, 0.25, and 0.5 mg/week) or microparticle-TiO2 (0.5 mg/week) by nonsurgical intratracheal instillation for 4 weeks. The results showed that Nano-TiO2 inhalation increased renal pathological changes in a dose-dependent manner. No renal pathological changes were observed in microparticle-TiO2-instilled mice. Nano-TiO2 (0.5 mg/week) possessed the ability to precipitate in the kidneys, determined by transmission electron microscopy and increased serum levels of blood urea nitrogen. The expressions of markers of ROS/RNS and renal fibrosis markers, including nitrotyrosine, inducible nitric oxide synthase, hypoxia inducible factor-1α (HIF-1α), heme oxygenase 1, transforming growth factor-β (TGFβ), and collagen I, determined by immunohistochemical staining were increased in the kidneys. Furthermore, Nano-TiO2-induced renal injury could be mitigated by iNOS inhibitor aminoguanidine and ROS scavenger N-acetylcysteine treatment in transcription level. The in vitro experiments showed that Nano-TiO2 significantly and dose-dependently increased the ROS production and the expressions of HIF-1α and TGFβ in human renal proximal tubular cells, which could be reversed by N-acetylcysteine treatment. Taken together, these results suggest Nano-TiO2 inhalation might induce renal fibrosis through a ROS/RNS-related HIF-1α-upregulated TGF-β signaling pathway.
Collapse
Affiliation(s)
- Kuo-Tong Huang
- Institute of Toxicology, College of Medicine, National Taiwan University , Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Wilker EH, Ljungman PL, Rice MB, Kloog I, Schwartz J, Gold DR, Koutrakis P, Vita JA, Mitchell GF, Vasan RS, Benjamin EJ, Hamburg NM, Mittleman MA. Relation of long-term exposure to air pollution to brachial artery flow-mediated dilation and reactive hyperemia. Am J Cardiol 2014; 113:2057-63. [PMID: 24793676 DOI: 10.1016/j.amjcard.2014.03.048] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 03/20/2014] [Accepted: 03/20/2014] [Indexed: 11/15/2022]
Abstract
Long-term exposure to ambient air pollution has been associated with cardiovascular morbidity and mortality. Impaired vascular responses may, in part, explain these findings, but the association of such long-term exposure with measures of both conduit artery and microvascular function has not been widely reported. We evaluated the association between residential proximity to a major roadway (primary or secondary highway) and spatially resolved average fine particulate matter (PM2.5) and baseline brachial artery diameter and mean flow velocity, flow-mediated dilation%, and hyperemic flow velocity, in the Framingham Offspring and Third Generation Cohorts. We examined 5,112 participants (2,731 [53%] women, mean age 49 ± 14 years). Spatially resolved average PM2.5 was associated with lower flow-mediated dilation% and hyperemic flow velocity. An interquartile range difference in PM2.5 (1.99 μg/m(3)) was associated with -0.16% (95% confidence interval [CI] -0.27%, -0.05%) lower flow-mediated dilation% and -0.72 (95% CI -1.38, -0.06) cm/s lower hyperemic flow velocity%. Residential proximity to a major roadway was negatively associated with flow-mediated dilation%. Compared with living ≥400 m away, living <50 m from a major roadway was associated with 0.32% lower flow-mediated dilation (95% CI -0.58%, -0.06%), but results for hyperemic flow velocity had wide confidence intervals -0.68 cm/s (95% CI -2.29, 0.93). In conclusion, residential proximity to a major roadway and higher levels of spatially resolved estimates of PM2.5 at participant residences are associated with impaired conduit artery and microvascular function in this large community-based cohort of middle-aged and elderly adults.
Collapse
Affiliation(s)
- Elissa H Wilker
- Cardiovascular Epidemiology Research Unit, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts; Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts.
| | - Petter L Ljungman
- Cardiovascular Epidemiology Research Unit, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts; Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Mary B Rice
- Cardiovascular Epidemiology Research Unit, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts; Pulmonary and Critical Care Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Itai Kloog
- Department of Geography and Environmental Development, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Joel Schwartz
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts
| | - Diane R Gold
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts
| | - Petros Koutrakis
- Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts
| | - Joseph A Vita
- NHLBI and Boston University's Framingham Heart Study, Framingham, Massachusetts; Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | | | - Ramachandran S Vasan
- NHLBI and Boston University's Framingham Heart Study, Framingham, Massachusetts; Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts; Sections of Preventive Medicine and Epidemiology and Cardiology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Emelia J Benjamin
- NHLBI and Boston University's Framingham Heart Study, Framingham, Massachusetts; Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Naomi M Hamburg
- NHLBI and Boston University's Framingham Heart Study, Framingham, Massachusetts; Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Murray A Mittleman
- Cardiovascular Epidemiology Research Unit, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts; Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts
| |
Collapse
|