1
|
Teixeira MI, Lopes CM, Amaral MH, Costa PC. Navigating Neurotoxicity and Safety Assessment of Nanocarriers for Brain Delivery: Strategies and Insights. Acta Biomater 2024:S1742-7061(24)00543-9. [PMID: 39307261 DOI: 10.1016/j.actbio.2024.09.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/02/2024] [Accepted: 09/17/2024] [Indexed: 10/11/2024]
Abstract
Nanomedicine, an area that uses nanomaterials for theragnostic purposes, is advancing rapidly, particularly in the detection and treatment of neurodegenerative diseases. The design of nanocarriers can be optimized to enhance drug bioavailability and targeting to specific organs, improving therapeutic outcomes. However, clinical translation hinges on biocompatibility and safety. Nanocarriers can cross the blood-brain barrier (BBB), potentially causing neurotoxic effects through mechanisms such as oxidative stress, DNA damage, and neuroinflammation. Concerns about their accumulation and persistence in the brain make it imperative to carry out a nanotoxicological risk assessment. Generally, this involves identifying exposure sources and routes, characterizing physicochemical properties, and conducting cytotoxicity assays both in vitro and in vivo. The lack of a specialized regulatory framework creates substantial gaps, making it challenging to translate findings across development stages. Additionally, there is a pressing need for innovative testing methods due to constraints on animal use and the demand for high-throughput screening. This review examines the mechanisms of nanocarrier-induced neurotoxicity and the challenges in risk assessment, highlighting the impact of physicochemical properties and the advantages and limitations of current neurotoxicity evaluation models. Future perspectives are also discussed. Additional guidance is crucial to improve the safety of nanomaterials and reduce associated uncertainty. STATEMENT OF SIGNIFICANCE: Nanocarriers show tremendous potential for theragnostic purposes in neurological diseases, enhancing drug targeting to the brain, and improving biodistribution and pharmacokinetics. However, their neurotoxicity is still a major field to be explored, with only 5% of nanotechnology-related publications addressing this matter. This review focuses on the issue of neurotoxicity and safety assessment of nanocarriers for brain delivery. Neurotoxicity-relevant exposure sources, routes, and molecular mechanisms, along with the impact of the physicochemical properties of nanomaterials, are comprehensively described. Moreover, the different experimental models used for neurotoxicity evaluation are explored at length, including their main advantages and limitations. To conclude, we discuss current challenges and future perspectives for a better understanding of risk assessment of nanocarriers for neurobiomedical applications.
Collapse
Affiliation(s)
- Maria Inês Teixeira
- UCIBIO - Applied Molecular Biosciences Unit, MedTech - Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Carla M Lopes
- UCIBIO - Applied Molecular Biosciences Unit, MedTech - Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; FP-I3ID, FP-ENAS/CEBIMED, Fernando Pessoa Energy, Environment, and Health Research Unit/Biomedical Research Center, Faculty of Health Sciences, Fernando Pessoa University, 4200-150 Porto, Portugal.
| | - Maria Helena Amaral
- UCIBIO - Applied Molecular Biosciences Unit, MedTech - Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Paulo C Costa
- UCIBIO - Applied Molecular Biosciences Unit, MedTech - Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| |
Collapse
|
2
|
Teglas T, Torices S, Taylor M, Coker D, Toborek M. Exposure to polychlorinated biphenyls selectively dysregulates endothelial circadian clock and endothelial toxicity. JOURNAL OF HAZARDOUS MATERIALS 2023; 454:131499. [PMID: 37126901 PMCID: PMC10202419 DOI: 10.1016/j.jhazmat.2023.131499] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/03/2023]
Abstract
Polychlorinated biphenyls (PCBs) are lipophilic and persistent environmental toxicants, which pose health threats to the exposed population. Among several organs and cell types, vascular tissue and endothelial cells are especially prone to PCB-induced toxicity. Exposure to PCBs can exert detrimental impacts on biological pathways, expression of transcription factors, and tight junction proteins that are integral to the functionality of endothelial cells. Because biological and cellular processes are tightly regulated by circadian rhythms, and disruption of the circadian system may cause several diseases, we evaluated if exposure to PCBs can alter the expression of the major endothelial circadian regulators. In addition, we studied if dysregulation of circadian rhythms by silencing the brain and muscle ARNT-like 1 (Bmal1) gene can contribute to alterations of brain endothelial cells in response to PCB treatment. We demonstrated that diminished expression of Bmal1 enhances PCB-induced dysregulation of tight junction complexes, such as the expression of occludin, JAM-2, ZO-1, and ZO-2 especially at pathologically relevant longer PCB exposure times. Overall, the obtained results imply that dysregulation of the circadian clock is involved in endothelial toxicity of PCBs. The findings provide new insights for toxicological studies focused on the interactions between environmental pollutants and regulation of circadian rhythms.
Collapse
Affiliation(s)
- Timea Teglas
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 33136, USA
| | - Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 33136, USA
| | - Madison Taylor
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 33136, USA
| | - Desiree Coker
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 33136, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 528E Gautier Bldg. 1011 NW 15th Street, Miami, FL 33136, USA; Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, Katowice, Poland.
| |
Collapse
|
3
|
Chen YP, Chou CM, Chang TY, Ting H, Dembélé J, Chu YT, Liu TP, Changou CA, Liu CW, Chen CT. Bridging Size and Charge Effects of Mesoporous Silica Nanoparticles for Crossing the Blood–Brain Barrier. Front Chem 2022; 10:931584. [PMID: 35880111 PMCID: PMC9307501 DOI: 10.3389/fchem.2022.931584] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/12/2022] [Indexed: 12/05/2022] Open
Abstract
The blood–brain barrier (BBB) is a highly selective cellular barrier that tightly controls the microenvironment of the central nervous system to restrict the passage of substances, which is a primary challenge in delivering therapeutic drugs to treat brain diseases. This study aimed to develop simple surface modifications of mesoporous silica nanoparticles (MSNs) without external stimuli or receptor protein conjugation, which exhibited a critical surface charge and size allowing them to cross the BBB. A series of MSNs with various charges and two different sizes of 50 and 200 nm were synthesized, which showed a uniform mesoporous structure with various surface zeta potentials ranging from +42.3 to −51.6 mV. Confocal microscopic results showed that 50 nm of strongly negatively charged N4-RMSN50@PEG/THPMP (∼−40 mV) could be significantly observed outside blood vessels of the brain in Tg(zfli1:EGFP) transgenic zebrafish embryos superior to the other negatively charged MSNs. However, very few positively charged MSNs were found in the brain, indicating that negatively charged MSNs could successfully penetrate the BBB. The data were confirmed by high-resolution images of 3D deconvoluted confocal microscopy and two-photon microscopy and zebrafish brain tissue sections. In addition, while increasing the size to 200 nm but maintaining the similar negative charge (∼40 mV), MSNs could not be detected in the brain of zebrafish, suggesting that transport across the BBB based on MSNs occurred in charge- and size-dependent manners. No obvious cytotoxicity was observed in the CTX-TNA2 astrocyte cell line and U87-MG glioma cell line treated with MSNs. After doxorubicin (Dox) loading, N4-RMSN50@PEG/THPMP/Dox enabled drug delivery and pH-responsive release. The toxicity assay showed that N4-RMSN50@PEG/THPMP could reduce Dox release, resulting in the increase of the survival rate in zebrafish. Flow cytometry demonstrated N4-RMSN50@PEG/THPMP had few cellular uptakes. Protein corona analysis revealed three transporter proteins, such as afamin, apolipoprotein E, and basigin, could contribute to BBB penetration, validating the possible mechanism of N4-RMSN50@PEG/THPMP crossing the BBB. With this simple approach, MSNs with critical negative charge and size could overcome the BBB-limiting characteristics of therapeutic drug molecules; furthermore, their use may also cause drug sustained-release in the brain, decreasing peripheral toxicity.
Collapse
Affiliation(s)
- Yi-Ping Chen
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Chih-Ming Chou
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry and Molecular Cell Biology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tsu-Yuan Chang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry and Molecular Cell Biology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hao Ting
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry and Molecular Cell Biology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Julien Dembélé
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - You-Tai Chu
- Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Tsang-Pai Liu
- Department of Surgery, Mackay Memorial Hospital, Taipei, Taiwan
| | - Chun A. Changou
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chien-Wei Liu
- Department of Information Management, St. Mary’s Junior College of Medicine, Nursing and Management, Yilan, Taiwan
| | - Chien-Tsu Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry and Molecular Cell Biology, College of Medicine, Taipei Medical University, Taipei, Taiwan
- *Correspondence: Chien-Tsu Chen,
| |
Collapse
|
4
|
Denuzière A, Ghersi-Egea JF. Cerebral concentration and toxicity of endocrine disrupting chemicals: The implication of blood-brain interfaces. Neurotoxicology 2022; 91:100-118. [DOI: 10.1016/j.neuro.2022.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/06/2022] [Accepted: 04/12/2022] [Indexed: 11/28/2022]
|
5
|
Nanosafety vs. nanotoxicology: adequate animal models for testing in vivo toxicity of nanoparticles. Toxicology 2021; 462:152952. [PMID: 34543703 DOI: 10.1016/j.tox.2021.152952] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/10/2021] [Accepted: 09/11/2021] [Indexed: 11/20/2022]
Abstract
Nanotoxicological studies using existing models of normal cells and animals often encounter a paradox: retention of nanoparticles in intracellular compartments for a long time is not accompanied by any significant toxicological effects. Can we expect that the revealed changes will be not harmful after translation to practice, outside of a sterile laboratory and ideally healthy organisms? Age-associated and pathological processes can affect target organs, metabolism, and detoxification in the mononuclear phagocyte system organs and change biodistribution routes, thus making the use of nanomaterial not safe. The potential solution to this issue can be testing the toxic properties of nanoparticles in animal models with chronic diseases. However, current studies of nanotoxicity in animal models with a brain, cardiovascular system, liver, digestive tract, reproductive system, and skin diseases are unsystematic. Even though these studies demonstrate the emergence of new toxic effects that are not present in healthy animals. In this regard, we set the goal of this review as the formulation of the requirements for an animal model capable of assessing the potential toxicity of nanoparticles based on the nanosafety approach.
Collapse
|
6
|
Murati T, Miletić M, Pleadin J, Šimić B, Kmetič I. Cell membrane-related toxic responses and disruption of intercellular communication in PCB mechanisms of toxicity: A review. J Appl Toxicol 2020; 40:1592-1601. [PMID: 32648282 DOI: 10.1002/jat.4019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/13/2020] [Accepted: 05/15/2020] [Indexed: 02/06/2023]
Abstract
An understanding of polychlorinated biphenyl (PCB) congener-specific effects on cell membrane and intercellular communication is important within the studies of PCB absorption, organ-related PCB accumulation and exertion of toxic responses. Toxic potential of PCBs is linked to various deleterious effects on human health, including neurotoxicity, immunotoxicity, reproductive toxicity and genotoxicity and, recently in 2016 International Agency for Research on Cancer (IARC) has upgraded the classification of PCBs to Group 1 "Carcinogenic to humans." Proposed mechanisms of aforementioned PCBs adverse effects at cellular membrane level are: (i) downregulation of gap junction intercellular communication and/or connexins; (ii) compromised membrane integrity; and (iii) altered tight junction barrier function. This study, based on an extensive literature survey, shows the progress in scientific research of each of these three levels with the aim of pointing out the earliest toxic events of PCBs, which can result in serious cell/tissue/organ damage.
Collapse
Affiliation(s)
- Teuta Murati
- Laboratory for Toxicology, Faculty of Food Technology and Biotechnology, University of Zagreb, Zagreb, Croatia
| | - Marina Miletić
- Laboratory for Toxicology, Faculty of Food Technology and Biotechnology, University of Zagreb, Zagreb, Croatia
| | - Jelka Pleadin
- Laboratory for Analytical Chemistry, Department of Veterinary Public Health, Croatian Veterinary Institute, Zagreb, Croatia
| | - Branimir Šimić
- Laboratory for Toxicology, Faculty of Food Technology and Biotechnology, University of Zagreb, Zagreb, Croatia
| | - Ivana Kmetič
- Laboratory for Toxicology, Faculty of Food Technology and Biotechnology, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
7
|
Ross AM, Mc Nulty D, O'Dwyer C, Grabrucker AM, Cronin P, Mulvihill JJ. Standardization of research methods employed in assessing the interaction between metallic-based nanoparticles and the blood-brain barrier: Present and future perspectives. J Control Release 2019; 296:202-224. [DOI: 10.1016/j.jconrel.2019.01.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 01/31/2023]
|
8
|
Harazin A, Bocsik A, Barna L, Kincses A, Váradi J, Fenyvesi F, Tubak V, Deli MA, Vecsernyés M. Protection of cultured brain endothelial cells from cytokine-induced damage by α-melanocyte stimulating hormone. PeerJ 2018; 6:e4774. [PMID: 29780671 PMCID: PMC5958884 DOI: 10.7717/peerj.4774] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 04/23/2018] [Indexed: 12/17/2022] Open
Abstract
The blood–brain barrier (BBB), an interface between the systemic circulation and the nervous system, can be a target of cytokines in inflammatory conditions. Pro-inflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) induce damage in brain endothelial cells and BBB dysfunction which contribute to neuronal injury. The neuroprotective effects of α-melanocyte stimulating hormone (α-MSH) were investigated in experimental models, but there are no data related to the BBB. Based on our recent study, in which α-MSH reduced barrier dysfunction in human intestinal epithelial cells induced by TNF-α and IL-1β, we hypothesized a protective effect of α-MSH on brain endothelial cells. We examined the effect of these two pro-inflammatory cytokines, and the neuropeptide α-MSH on a culture model of the BBB, primary rat brain endothelial cells co-cultured with rat brain pericytes and glial cells. We demonstrated the expression of melanocortin-1 receptor in isolated rat brain microvessels and cultured brain endothelial cells by RT-PCR and immunohistochemistry. TNF-α and IL-1β induced cell damage, measured by impedance and MTT assay, which was attenuated by α-MSH (1 and 10 pM). The peptide inhibited the cytokine-induced increase in brain endothelial permeability, and restored the morphological changes in cellular junctions visualized by immunostaining for claudin-5 and β-catenin. Elevated production of reactive oxygen species and the nuclear translocation of NF-κB were also reduced by α-MSH in brain endothelial cells stimulated by cytokines. We demonstrated for the first time the direct beneficial effect of α-MSH on cultured brain endothelial cells, indicating that this neurohormone may be protective at the BBB.
Collapse
Affiliation(s)
- András Harazin
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.,Doctoral School in Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Alexandra Bocsik
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Lilla Barna
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.,Doctoral School in Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - András Kincses
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Judit Váradi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Ferenc Fenyvesi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | | | - Maria A Deli
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Miklós Vecsernyés
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
9
|
Long Y, Huang C, Wu J, Cheng JN, Liang GN, Jiang CX, Wan Q. 2,3',4,4',5-Pentachlorobiphenyl impairs insulin-induced NO production partly through excessive ROS production in endothelial cells. Toxicol Mech Methods 2017; 27:592-597. [PMID: 28592194 DOI: 10.1080/15376516.2017.1337259] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Polychlorinated biphenyls (PCBs) have been reported to be associated with increased risk to hypertension, atherosclerosis, cardiovascular disease, etc. 2,3',4,4',5-Pentachlorobiphenyl, known as PCB-118, is a member of coplanar PCBs which renders their structure similar to polychlorinated dibenzo-p-dioxins (PCDDs) and has dioxin-like activity. In our current study, we investigated the effect of PCB-118 exposure on nitric oxide (NO) production and the underlying mechanisms in vitro. Exposure of PCB-118 impaired insulin-induced NO production and endothelial nitric oxide synthase (eNOS) activity in human umbilical vein endothelial cells (HUVECs) with no significant effect on cell viability. Furthermore, PCB-118 treatment induced oxidative stress. In addition, scavenging of reactive oxygen species (ROS) by 10 μM N-acetyl-l-cysteine (NAC) partly rescued impaired insulin-induced eNOS activities and NO productions induced by PCB-118 in HUVECs. Taken together, these results indicate that PCB-118 mediates lower eNOS activity and impairs insulin-induced NO production partly through excessive ROS production in endothelial cells.
Collapse
Affiliation(s)
- Yang Long
- a Department of Endocrinology , t he Affiliated Hospital of Southwest Medical University , Luzhou , Sichuan , PR China.,b Laboratory of Endocrinology , Experimental Medicine Center, the Affiliated Hospital of Southwest Medical University , Luzhou , Sichuan , PR China
| | - Can Huang
- b Laboratory of Endocrinology , Experimental Medicine Center, the Affiliated Hospital of Southwest Medical University , Luzhou , Sichuan , PR China
| | - Jian Wu
- b Laboratory of Endocrinology , Experimental Medicine Center, the Affiliated Hospital of Southwest Medical University , Luzhou , Sichuan , PR China
| | - Jin-Nan Cheng
- c Department of Dermatology , t he Affiliated Hospital of Southwest Medical University , Luzhou , Sichuan , PR China
| | - Guan-Nan Liang
- b Laboratory of Endocrinology , Experimental Medicine Center, the Affiliated Hospital of Southwest Medical University , Luzhou , Sichuan , PR China
| | - Chun-Xia Jiang
- a Department of Endocrinology , t he Affiliated Hospital of Southwest Medical University , Luzhou , Sichuan , PR China.,b Laboratory of Endocrinology , Experimental Medicine Center, the Affiliated Hospital of Southwest Medical University , Luzhou , Sichuan , PR China
| | - Qin Wan
- a Department of Endocrinology , t he Affiliated Hospital of Southwest Medical University , Luzhou , Sichuan , PR China
| |
Collapse
|
10
|
Blood-brain barrier dysfunction induced by silica NPs in vitro and in vivo : Involvement of oxidative stress and Rho-kinase/JNK signaling pathways. Biomaterials 2017; 121:64-82. [DOI: 10.1016/j.biomaterials.2017.01.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 01/03/2017] [Accepted: 01/03/2017] [Indexed: 01/03/2023]
|
11
|
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: 5.5] [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
|
12
|
Eum SY, Jaraki D, András IE, Toborek M. Lipid rafts regulate PCB153-induced disruption of occludin and brain endothelial barrier function through protein phosphatase 2A and matrix metalloproteinase-2. Toxicol Appl Pharmacol 2015; 287:258-66. [PMID: 26080028 DOI: 10.1016/j.taap.2015.06.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 05/20/2015] [Accepted: 06/11/2015] [Indexed: 01/30/2023]
Abstract
Occludin is an essential integral transmembrane protein regulating tight junction (TJ) integrity in brain endothelial cells. Phosphorylation of occludin is associated with its localization to TJ sites and incorporation into intact TJ assembly. The present study is focused on the role of lipid rafts in polychlorinated biphenyl (PCB)-induced disruption of occludin and endothelial barrier function. Exposure of human brain endothelial cells to 2,2',4,4',5,5'-hexachlorobiphenyl (PCB153) induced dephosphorylation of threonine residues of occludin and displacement of occludin from detergent-resistant membrane (DRM)/lipid raft fractions within 1h. Moreover, lipid rafts modulated the reduction of occludin level through activation of matrix metalloproteinase 2 (MMP-2) after 24h PCB153 treatment. Inhibition of protein phosphatase 2A (PP2A) activity by okadaic acid or fostriecin markedly protected against PCB153-induced displacement of occludin and increased permeability of endothelial cells. The implication of lipid rafts and PP2A signaling in these processes was further defined by co-immunoprecipitation of occludin with PP2A and caveolin-1, a marker protein of lipid rafts. Indeed, a significant MMP-2 activity was observed in lipid rafts and was increased by exposure to PCB153. The pretreatment of MMP-2 inhibitors protected against PCB153-induced loss of occludin and disruption of lipid raft structure prevented the increase of endothelial permeability. Overall, these results indicate that lipid raft-associated processes, such as PP2A and MMP-2 activation, participate in PCB153-induced disruption of occludin function in brain endothelial barrier. This study contributes to a better understanding of the mechanisms leading to brain endothelial barrier dysfunction in response to exposure to environmental pollutants, such as ortho-substituted PCBs.
Collapse
Affiliation(s)
- Sung Yong Eum
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Dima Jaraki
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ibolya E András
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Michal Toborek
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
13
|
Hendryx M, Luo J, Chen BC. Total and cardiovascular mortality rates in relation to discharges from Toxics Release Inventory sites in the United States. ENVIRONMENTAL RESEARCH 2014; 133:36-41. [PMID: 24906066 DOI: 10.1016/j.envres.2014.05.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 04/28/2014] [Accepted: 05/02/2014] [Indexed: 06/03/2023]
Abstract
BACKGROUND This study analyzed Toxics Release Inventory (TRI) discharges in association with covariate-adjusted total and cardiovascular mortality rates for males and females in US counties. METHODS Average annual county-level reported releases from TRI facilities measured in pounds per square mile which were calculated for the years 1990-1999, and tested for associations with age-adjusted mortality rates for 2006-2010. Chemicals were grouped into four categories: 1) carcinogens, 2) metals, 3) hazardous air pollutants, and 4) chemicals in the Comprehensive Environmental Response, Compensation and Liability Act. For each of these chemical groups the reported total, water, and air emissions were measured. Age-adjusted mortality rates were found separately for males and females from the Centers for Disease Control and Prevention for total and cardiovascular disease. Covariates included rates of smoking, obesity, high school and college education, race/ethnicity, poverty, unemployment, percent without health insurance, and urban-rural setting. Data were analyzed using multiple linear regression models. RESULTS Greater average annual TRI releases in 1990-1999 in all four chemical categories were significantly associated with higher mortality rates in 2006-2010 for both total and cardiovascular mortality, and for both males and females, adjusted for covariates. Associations were stronger for air releases than for water releases. CONCLUSIONS This study provides the first evidence that greater amounts of TRI releases are related to higher population mortality rates for cardiovascular disease. In addition, the study showed that adverse TRI effects were broadly present for both males and females for multiple chemical groups. Further progress is needed to reduce the use and release of harmful chemicals from TRI facilities in the United States.
Collapse
Affiliation(s)
- Michael Hendryx
- Department of Applied Health Science, School of Public Health, Indiana University, Bloomington, IN, USA.
| | - Juhua Luo
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University, Bloomington, IN, USA.
| | - Bo-Chiuan Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
14
|
Fabulas-da Costa A, Aijjou R, Hachani J, Landry C, Cecchelli R, Culot M. In vitro blood–brain barrier model adapted to repeated-dose toxicological screening. Toxicol In Vitro 2013; 27:1944-53. [DOI: 10.1016/j.tiv.2013.06.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 06/14/2013] [Accepted: 06/26/2013] [Indexed: 11/28/2022]
|
15
|
TLR4 signaling is involved in brain vascular toxicity of PCB153 bound to nanoparticles. PLoS One 2013; 8:e63159. [PMID: 23690990 PMCID: PMC3653967 DOI: 10.1371/journal.pone.0063159] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 03/29/2013] [Indexed: 11/26/2022] Open
Abstract
PCBs bind to environmental particles; however, potential toxicity exhibited by such complexes is not well understood. The aim of the present study is to study the hypothesis that assembling onto nanoparticles can influence the PCB153-induced brain endothelial toxicity via interaction with the toll-like receptor 4 (TLR4). To address this hypothesis, TLR4-deficient and wild type control mice (males, 10 week old) were exposed to PCB153 (5 ng/g body weight) bound to chemically inert silica nanoparticles (PCB153-NPs), PCB153 alone, silica nanoparticles (NPs; diameter, 20 nm), or vehicle. Selected animals were also subjected to 40 min ischemia, followed by a 24 h reperfusion. As compared to exposure to PCB153 alone, treatment with PCB153-NP potentiated the brain infarct volume in control mice. Importantly, this effect was attenuated in TLR4-deficient mice. Similarly, PCB153-NP-induced proinflammatory responses and disruption of tight junction integrity were less pronounced in TLR4-deficient mice as compared to control animals. Additional in vitro experiments revealed that TLR4 mediates toxicity of PCB153-NP via recruitment of tumor necrosis factor-associated factor 6 (TRAF6). The results of current study indicate that binding to seemingly inert nanoparticles increase cerebrovascular toxicity of PCBs and suggest that targeting the TLR4/TRAF6 signaling may protect against these effects.
Collapse
|