1
|
Aygörmez S, Küçükler S, Gür C, Akaras N, Maraşli Ş, Mehmet Kandemir F. Investigation of the effects of morin on potassium bromate-induced brain damage in rats via different pathways with biochemical and histopathological methods. Food Chem Toxicol 2025; 201:115466. [PMID: 40252904 DOI: 10.1016/j.fct.2025.115466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/25/2025] [Accepted: 04/17/2025] [Indexed: 04/21/2025]
Abstract
AIM Potassium bromate (KBrO3) is a colorless, odorless substance used as a food additive. It causes multiple organ damage and neurotoxicity. Morin is a flavonoid from the Moraceae family known to have anti-inflammatory, antioxidant, antiapoptotic, antiautophagic, and neuroprotective properties. Therefore, this study aimed to investigate the effects of Morin against KBrO3-induced brain damage. METHODS 62 mg/kg KBrO3 and 50-100 mg/kg Morin were administered to 35 male rats by oral gavage daily for 14 days. Various analyses were performed using molecular, biochemical, and histological methods. RESULTS The analyses results showed that KBrO3 application decreased antioxidant markers and raised lipid peroxidation in the brain tissue. The KBrO3 application triggered apoptosis, endoplasmic reticulum stress, and inflammation. Morin treatment increased enzymatic and nonenzymatic antioxidant levels and decreased lipid peroxidation. In addition, Morin alleviated KBrO3-induced apoptosis, endoplasmic reticulum stress, and inflammation in the brain tissue. The histopathological analysis revealed an increase in degenerative changes, as well as pyknotic changes and vacuolization in cells, in neurons in the KBrO3 group. Increased hyperemia and congestion were detected in the meninges and vessels in the cerebral cortex. CONCLUSION KBrO3 application caused toxicity in the brain tissue and impaired tissue integrity, whereas Morin treatment alleviated KBrO3-induced toxicity.
Collapse
Affiliation(s)
- Serpil Aygörmez
- Department of Veterinary Biochemistry, Faculty of Veterinary, Kafkas University, Kars, Turkey.
| | - Sefa Küçükler
- Department of Veterinary Biochemistry, Faculty of Veterinary, Atatürk University, Erzurum, Turkey
| | - Cihan Gür
- Department of Medical Laboratory Techniques, Vocational School of Health Services, Atatürk University, Erzurum, Turkey
| | - Nurhan Akaras
- Department of Histology and Embryology, Faculty of Medicine, Aksaray University, Aksaray, Turkey
| | - Şaban Maraşli
- Department of Veterinary Biochemistry, Faculty of Veterinary, Kafkas University, Kars, Turkey
| | - Fatih Mehmet Kandemir
- Department of Medical Biochemistry, Faculty of Medicine, Aksaray University, Aksaray, Turkey
| |
Collapse
|
2
|
Vysakh VG, Sukumaran S, Gopalakrishnan A. Evaluating the effects of zinc oxide nanoparticles on a sentinel aquatic invertebrate species: Transcriptomic analysis and potential implications for ecosystem health. MARINE POLLUTION BULLETIN 2025; 212:117570. [PMID: 39824139 DOI: 10.1016/j.marpolbul.2025.117570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 01/20/2025]
Abstract
The widespread use of zinc oxide nanoparticles (ZnO NPs) in various products raises significant ecological concerns due to their potential toxic effects in aquatic environments. This study employed the Asian green mussel (Perna viridis) as a model to explore the molecular and ecological risks of ZnO NP exposure using transcriptomics. Mussels exposed to ZnO NPs (5, 10, and 15 mg/L) for 28 days showed significant gene expression changes in gill tissues, affecting immune response, calcium homeostasis, and cellular stress. Disrupted pathways such as FOXO, Wnt, and TGFβ reveal complex toxicity mechanisms. These findings provide crucial insights into the environmental impact of nanoparticle pollution, emphasizing the need for stringent regulations. Furthermore, the shared molecular pathways suggest that similar mechanisms may occur in humans, highlighting potential health risks associated with nanoparticle exposure.
Collapse
Affiliation(s)
- V G Vysakh
- Marine Biotechnology Fish Nutrition and Health Division, Central Marine Fisheries Research Institute, Post Box No 1603, Ernakulam North PO., Kochi 682018, Kerala, India; Mangalore University. Mangalagangotri, Mangalore 574199, Karnataka, India
| | - Sandhya Sukumaran
- Marine Biotechnology Fish Nutrition and Health Division, Central Marine Fisheries Research Institute, Post Box No 1603, Ernakulam North PO., Kochi 682018, Kerala, India.
| | - A Gopalakrishnan
- Marine Biotechnology Fish Nutrition and Health Division, Central Marine Fisheries Research Institute, Post Box No 1603, Ernakulam North PO., Kochi 682018, Kerala, India
| |
Collapse
|
3
|
Xiao P, Wu S, Wang Z, Shen G, Shi X. Biotoxicity of paraquat to lung cells mediated by endoplasmic reticulum-mitochondria interaction. J Mol Histol 2024; 55:1063-1077. [PMID: 39215928 DOI: 10.1007/s10735-024-10249-7] [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: 06/10/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
The high lethality caused by paraquat (PQ) poisoning has attracted much attention in public and human health due to its high toxicity and lethality. However, the understanding of the mechanism of PQ-induced apoptosis from the perspective of organelles, especially inter-organelle interactions, is still scarce. Exploring the linkage of multiple organelles during PQ poisoning and the molecular mechanisms of PQ poisoning under its mediation will help to gain insight into the mode of PQ poisoning at the organelle level. In this study, we observed that a certain dose of PQ gavage induced oxidative stress, mitochondrial dysfunction and endoplasmic reticulum stress in rat lung tissue cells. PQ toxicity led to the occurrence of Ca2+ overload in the endoplasmic reticulum, and the activated BIP and CHOP pathways directly/indirectly led to the expression of apoptogenic factors Caspase family factors. In addition, PQ promoted Ca2+ release from the endoplasmic reticulum and Ca2+ uptake by mitochondria, which induced the disruption of Bax/Bcl-2 channel proteins in response to the IP3R/RyR/VDAC1&2/MCU Ca2+ axis thereby leading to the release of CytoC, which ultimately induced endoplasmic reticulum stress and apoptotic cell death. In addition, 10 differential proteins were screened and validated by proteomics that may act as upstream and downstream active factors of mitochondria-endoplasmic reticulum interaction-mediated biotoxicity. Our findings provide new perspectives for researchers to explore the toxicity mechanisms of PQ to reduce their adverse effects.
Collapse
Affiliation(s)
- Ping Xiao
- Clinical Laboratory, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Shaohua Wu
- Clinical Laboratory, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Zhiyong Wang
- Department of Emergency, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Guoqiang Shen
- Department of Emergency, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Xiaofeng Shi
- Department of Emergency, Tianjin First Central Hospital, Tianjin, 300192, China.
| |
Collapse
|
4
|
Singh G, Thakur N, Kumar R. Nanoparticles in drinking water: Assessing health risks and regulatory challenges. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 949:174940. [PMID: 39047836 DOI: 10.1016/j.scitotenv.2024.174940] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
Nanoparticles (NPs) pose a significant concern in drinking water due to their potential health risks and environmental impact. This review provides a comprehensive analysis of the current understanding of NP sources and contamination in drinking water, focusing on health concerns, mitigation strategies, regulatory frameworks, and future perspectives. This review highlights the importance of nano-specific pathways, fate processes, health risks & toxicity, and the need for realistic toxicity assessments. Different NPs like titanium dioxide, silver, nanoplastics, nanoscale liquid crystal monomers, copper oxide, and others pose potential health risks through ingestion, inhalation, or dermal exposure, impacting organs and potentially leading to oxidative stress, inflammatory responses, DNA damage, cytotoxicity, disrupt intracellular energetic mechanisms, reactive oxygen species generation, respiratory and immune toxicity, and genotoxicity in humans. Utilizing case studies and literature reviews, we investigate the health risks associated with NPs in freshwater environments, emphasizing their relevance to drinking water quality. Various mitigation and treatment strategies, including filtration systems (e.g., reverse osmosis, and ultra/nano-filtration), adsorption processes, coagulation/flocculation, electrocoagulation, advanced oxidation processes, membrane distillation, and ultraviolet treatment, all of which demonstrate high removal efficiencies for NPs from drinking water. Regulatory frameworks and challenges for the production, applications, and disposal of NPs at both national and international levels are discussed, emphasizing the need for tailored regulations to address NP contamination and standardize safety testing and risk assessment practices. Looking ahead, this review underscores the necessity of advancing detection methods and nanomaterial-based treatment technologies while stressing the pivotal role of public awareness and tailored regulatory guidelines in upholding drinking water quality standards. This review emphasizes the urgency of addressing NP contamination in drinking water and provides insights into potential solutions and future research directions. Lastly, this review worth concluded with future recommendations on advanced analytical techniques and sensitive sensors for NP detection for safeguarding public health and policy implementations.
Collapse
Affiliation(s)
- Gagandeep Singh
- Department of Biosciences (UIBT), Chandigarh University, Ludhiana, Punjab 140413, India
| | - Neelam Thakur
- Department of Zoology, Sardar Patel University, Vallabh Government College, Campus, Mandi, Himachal Pradesh 175001, India.
| | - Rakesh Kumar
- Department of Biosystems Engineering, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|
5
|
Yan KX, Ge BJ, Sang R, Zhao P, Liu XM, Yu MH, Liu XT, Qiu Q, Zhang XM. Taraxasterol attenuates zearalenone-induced kidney damage in mice by modulating oxidative stress and endoplasmic reticulum stress. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117093. [PMID: 39317070 DOI: 10.1016/j.ecoenv.2024.117093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/14/2024] [Accepted: 09/20/2024] [Indexed: 09/26/2024]
Abstract
Taraxasterol is one of the bioactive ingredients from traditional Chinese herb Taraxacum, which exhibits multiple pharmacological activities and protective effects. However, the underlying influence and mechanism of its use against kidney damage caused from zearalenone (ZEA) remain unexplored. The ZEA-induced kidney damage model of mice was established by feeding diets containing ZEA (2 mg/kg), and taraxasterol (5 and 10 mg/kg) was administered by gavage for 28 days. Results demonstrated taraxasterol increased average daily gain (ADG) and average daily feed intake (ADFI), reduced feed-to-gain ratio (F/G) and kidney index of mice induced by ZEA. Taraxasterol alleviated histopathological changes of kidney, reduced ZEA residue and the levels of blood urea nitrogen (BUN), uric acid (UA), and creatinine (CRE). Concurrently, taraxasterol reduced the contents of oxidative stress indicator reactive oxygen species (ROS) and malondialdehyde (MDA), and increased the activities of antioxidant enzymes catalase (CAT), total superoxide dismutase (T-SOD), and glutathione peroxidase (GSH-Px). Further, taraxasterol up-regulated the mRNA and protein expression of nuclear factor erythroid-2-related factor 2 (Nrf2), GSH-Px, NAD(P)H quinone oxidoreductase 1 (NQO1), and heme oxygenase-1 (HO-1), and down-regulated the mRNA and protein expression of KELCH like ECH associated protein (Keap1) in Nrf2/Keap1 pathway. Taraxasterol down-regulated the mRNA and protein expression of immunoglobulin binding protein (Bip), C/EBP homologous protein (CHOP), Bcl-2 associated X (Bax), cysteine protease (Caspase)-12, and Caspase-3, and up-regulated B-cell lymphoma 2 (Bcl-2) expression in endoplasmic reticulum stress pathway. This study suggests that taraxasterol attenuates ZEA-induced mouse kidney damage through the modulation of Nrf2/Keapl pathway to play antioxidant role and endoplasmic reticulum stress pathway to enhance anti-apoptotic ability. It will provide a basis for taraxasterol as a potential drug to prevent and treat ZEA-induced kidney damage.
Collapse
Affiliation(s)
- Ke-Xin Yan
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Gongyuan Street, Yanji, Jilin 133000, China.
| | - Bing-Jie Ge
- College of Agriculture, Yanbian University, Gongyuan Street, Yanji, Jilin 133000, China.
| | - Rui Sang
- College of Agriculture, Yanbian University, Gongyuan Street, Yanji, Jilin 133000, China.
| | - Peng Zhao
- College of Agriculture, Yanbian University, Gongyuan Street, Yanji, Jilin 133000, China.
| | - Xin-Man Liu
- College of Agriculture, Yanbian University, Gongyuan Street, Yanji, Jilin 133000, China.
| | - Ming-Hong Yu
- College of Agriculture, Yanbian University, Gongyuan Street, Yanji, Jilin 133000, China.
| | - Xiao-Tong Liu
- College of Agriculture, Yanbian University, Gongyuan Street, Yanji, Jilin 133000, China.
| | - Qian Qiu
- College of Agriculture, Yanbian University, Gongyuan Street, Yanji, Jilin 133000, China.
| | - Xue-Mei Zhang
- College of Agriculture, Yanbian University, Gongyuan Street, Yanji, Jilin 133000, China.
| |
Collapse
|
6
|
Li LY, Park E, He C, Abbasi AZ, Henderson JT, Fraser PE, Uetrecht JP, Rauth AM, Wu XY. Evaluation of the biodistribution and preliminary safety profile of a novel brain-targeted manganese dioxide-based nanotheranostic system for Alzheimer's disease. Nanotoxicology 2024; 18:315-334. [PMID: 38847611 DOI: 10.1080/17435390.2024.2361687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 05/07/2024] [Accepted: 05/27/2024] [Indexed: 08/03/2024]
Abstract
A novel brain-targeted and reactive oxygen species-activatable manganese dioxide containing nanoparticle system functionalized with anti-amyloid-β antibody (named aAβ-BTRA-NC) developed by our group has shown great promise as a highly selective magnetic resonance imaging (MRI) contrast agent for early detection and multitargeted disease-modifying treatment of Alzheimer's disease (AD). To further evaluate the suitability of the formulation for future clinical application, we investigated the safety, biodistribution, and pharmacokinetic profile of aAβ-BTRA-NC in a transgenic TgCRND8 mouse AD model, wild type (WT) littermate, and CD-1 mice. Dose-ascending studies demonstrated that aAβ-BTRA-NC was well-tolerated by the animals up to 300 μmol Mn/kg body weight [b.w.], 3 times the efficacious dose for early AD detection without apparent adverse effects; Histopathological, hematological, and biochemical analyses indicated that a single dose of aAβ-BTRA-NC did not cause any toxicity in major organs. Immunotoxicity data showed that aAβ-BTRA-NC was safer than commercially available gadolinium-based MRI contrast agents at an equivalent dose of 100 μmol/kg b.w. of metal ions. Intravenously administered aAβ-BTRA-NC was taken up by main organs with the order of liver, kidneys, intestines, spleen, followed by other organs, and cleared after one day to one week post injection. Pharmacokinetic analysis indicated that the plasma concentration profile of aAβ-BTRA-NC followed a 2-compartmental model with faster clearance in the AD mice than in the WT mice. The results suggest that aAβ-BTRA-NC exhibits a strong safety profile as a nanotheranostic agent which warrants more robust preclinical development for future clinical applications.
Collapse
Affiliation(s)
- Lily Yi Li
- Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Elliya Park
- Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Chunsheng He
- Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Azhar Z Abbasi
- Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Jeffrey T Henderson
- Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Jack P Uetrecht
- Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Andrew M Rauth
- Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Xiao Yu Wu
- Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Das SK, Sen K, Ghosh B, Ghosh N, Sinha K, Sil PC. Molecular mechanism of nanomaterials induced liver injury: A review. World J Hepatol 2024; 16:566-600. [PMID: 38689743 PMCID: PMC11056894 DOI: 10.4254/wjh.v16.i4.566] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/05/2024] [Accepted: 03/19/2024] [Indexed: 04/24/2024] Open
Abstract
The unique physicochemical properties inherent to nanoscale materials have unveiled numerous potential applications, spanning beyond the pharmaceutical and medical sectors into various consumer industries like food and cosmetics. Consequently, humans encounter nanomaterials through diverse exposure routes, giving rise to potential health considerations. Noteworthy among these materials are silica and specific metallic nanoparticles, extensively utilized in consumer products, which have garnered substantial attention due to their propensity to accumulate and induce adverse effects in the liver. This review paper aims to provide an exhaustive examination of the molecular mechanisms underpinning nanomaterial-induced hepatotoxicity, drawing insights from both in vitro and in vivo studies. Primarily, the most frequently observed manifestations of toxicity following the exposure of cells or animal models to various nanomaterials involve the initiation of oxidative stress and inflammation. Additionally, we delve into the existing in vitro models employed for evaluating the hepatotoxic effects of nanomaterials, emphasizing the persistent endeavors to advance and bolster the reliability of these models for nanotoxicology research.
Collapse
Affiliation(s)
- Sanjib Kumar Das
- Department of Zoology, Jhargram Raj College, Jhargram 721507, India
| | - Koushik Sen
- Department of Zoology, Jhargram Raj College, Jhargram 721507, India
| | - Biswatosh Ghosh
- Department of Zoology, Bidhannagar College, Kolkata 700064, India
| | - Nabanita Ghosh
- Department of Zoology, Maulana Azad College, Kolkata 700013, India
| | - Krishnendu Sinha
- Department of Zoology, Jhargram Raj College, Jhargram 721507, India.
| | - Parames C Sil
- Department of Molecular Medicine, Bose Institute, Calcutta 700054, India
| |
Collapse
|
8
|
Moselhy WA, Ibrahim MA, Khalifa AG, El-Nahass ES, Hassan NEHY. The effects of TiO2, ZnO, IONs and Al2O3 metallic nanoparticles on the CYP1A1 and NBN transcripts in rat liver. Toxicol Res (Camb) 2024; 13:tfae034. [PMID: 38559758 PMCID: PMC10980790 DOI: 10.1093/toxres/tfae034] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/04/2024] [Accepted: 02/22/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction Metal oxide nanoparticles are currently used widely in many aspects of human and animal life with broad prospects for biomedical purposes. The present work was carried out to investigate the effects of orally administrated TiO2NPs, ZnONPs, IONs and Al2O3NPs on the mRNA expression level of CYP 1A1 and NBN in the rat liver. Materials and Methods Four groups of male Albino rats were given their respective treatment orally for 60 days in a dose of 1/20 of the LD50 TiO2NPs (600 mg/Kg b.wt/day), ZnONPs (340 mg/Kg b.wt/day), IONs (200 mg/kg b.wt/day) and Al2O3NPs (100 mg/Kg b.wt/day) and a fifth group served as a control group. Rresults The mRNA level of CYP 1A1 and NBN showed up-regulation in all the NPs-treated groups relative to the control group. ZnONPs group recorded the highest expression level while the TiO2NPs group showed the lowest expression level transcript. Conclusion:The toxic effects produced by these nanoparticles were more pronounced in the case of zinc oxide, followed by aluminum oxide, iron oxide nanoparticles and titanium dioxide, respectively.
Collapse
Affiliation(s)
- Walaa A Moselhy
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Marwa A Ibrahim
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, Cairo 12211, Egypt
| | - Ahlam G Khalifa
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - El-Shaymaa El-Nahass
- Department of Pathology, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Nour El-Houda Y Hassan
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
9
|
Vagena IA, Gatou MA, Theocharous G, Pantelis P, Gazouli M, Pippa N, Gorgoulis VG, Pavlatou EA, Lagopati N. Functionalized ZnO-Based Nanocomposites for Diverse Biological Applications: Current Trends and Future Perspectives. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:397. [PMID: 38470728 PMCID: PMC10933906 DOI: 10.3390/nano14050397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 02/18/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024]
Abstract
The wide array of structures and characteristics found in ZnO-based nanostructures offers them a versatile range of uses. Over the past decade, significant attention has been drawn to the possible applications of these materials in the biomedical field, owing to their distinctive electronic, optical, catalytic, and antimicrobial attributes, alongside their exceptional biocompatibility and surface chemistry. With environmental degradation and an aging population contributing to escalating healthcare needs and costs, particularly in developing nations, there's a growing demand for more effective and affordable biomedical devices with innovative functionalities. This review delves into particular essential facets of different synthetic approaches (chemical and green) that contribute to the production of effective multifunctional nano-ZnO particles for biomedical applications. Outlining the conjugation of ZnO nanoparticles highlights the enhancement of biomedical capacity while lowering toxicity. Additionally, recent progress in the study of ZnO-based nano-biomaterials tailored for biomedical purposes is explored, including biosensing, bioimaging, tissue regeneration, drug delivery, as well as vaccines and immunotherapy. The final section focuses on nano-ZnO particles' toxicity mechanism with special emphasis to their neurotoxic potential, as well as the primary toxicity pathways, providing an overall review of the up-to-date development and future perspectives of nano-ZnO particles in the biomedicine field.
Collapse
Affiliation(s)
- Ioanna-Aglaia Vagena
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National Kapodistrian University of Athens (NKUA), 11527 Athens, Greece; (I.-A.V.); (M.G.)
| | - Maria-Anna Gatou
- Laboratory of General Chemistry, School of Chemical Engineering, National Technical University of Athens, Zografou Campus, 15772 Athens, Greece; (M.-A.G.); (E.A.P.)
| | - Giorgos Theocharous
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National Kapodistrian University of Athens (NKUA), 11527 Athens, Greece; (G.T.); (P.P.)
| | - Pavlos Pantelis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National Kapodistrian University of Athens (NKUA), 11527 Athens, Greece; (G.T.); (P.P.)
| | - Maria Gazouli
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National Kapodistrian University of Athens (NKUA), 11527 Athens, Greece; (I.-A.V.); (M.G.)
- School of Science and Technology, Hellenic Open University, 26335 Patra, Greece
| | - Natassa Pippa
- Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National Kapodistrian University of Athens (NKUA), 15771 Athens, Greece;
| | - Vassilis G. Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National Kapodistrian University of Athens (NKUA), 11527 Athens, Greece; (G.T.); (P.P.)
- Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
- Ninewells Hospital and Medical School, University of Dundee, Dundee DD19SY, UK
- Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M20 4GJ, UK
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7YH, UK
| | - Evangelia A. Pavlatou
- Laboratory of General Chemistry, School of Chemical Engineering, National Technical University of Athens, Zografou Campus, 15772 Athens, Greece; (M.-A.G.); (E.A.P.)
| | - Nefeli Lagopati
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National Kapodistrian University of Athens (NKUA), 11527 Athens, Greece; (I.-A.V.); (M.G.)
- Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
10
|
Rajan SS, Chandran R, Abrahamse H. Overcoming challenges in cancer treatment: Nano-enabled photodynamic therapy as a viable solution. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1942. [PMID: 38456341 DOI: 10.1002/wnan.1942] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/09/2024] [Accepted: 01/17/2024] [Indexed: 03/09/2024]
Abstract
Cancer presents a formidable challenge, necessitating innovative therapies that maximize effectiveness while minimizing harm to healthy tissues. Nanotechnology has emerged as a transformative force in cancer treatment, particularly through nano-enabled photodynamic therapy (NE-PDT), which leverages precise and targeted interventions. NE-PDT capitalizes on photosensitizers activated by light to generate reactive oxygen species (ROS) that initiate apoptotic pathways in cancer cells. Nanoparticle enhancements optimize this process, improving drug delivery, selectivity, and ROS production within tumors. This review dissects NE-PDT's mechanistic framework, showcasing its potential to harness apoptosis as a potent tool in cancer therapy. Furthermore, the review explores the synergy between NE-PDT and complementary treatments like chemotherapy, immunotherapy, and targeted therapies, highlighting the potential to amplify apoptotic responses, enhance immune recognition of cancer cells, and inhibit resistance mechanisms. Preclinical and clinical advancements in NE-PDT demonstrate its efficacy across various cancer types. Challenges in translating NE-PDT into clinical practice are also addressed, emphasizing the need for optimizing nanoparticle design, refining dosimetry, and ensuring long-term safety. Ultimately, NE-PDT represents a promising approach in cancer therapy, utilizing the intricate mechanisms of apoptosis to address therapeutic hurdles. The review underscores the importance of understanding the interplay between nanoparticles, ROS generation, and apoptotic pathways, contributing to a deeper comprehension of cancer biology and novel therapeutic strategies. As interdisciplinary collaborations continue to thrive, NE-PDT offers hope for effective and targeted cancer interventions, where apoptosis manipulation becomes central to conquering cancer. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Sheeja S Rajan
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Rahul Chandran
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
11
|
Ding R, Li Y, Yu Y, Sun Z, Duan J. Prospects and hazards of silica nanoparticles: Biological impacts and implicated mechanisms. Biotechnol Adv 2023; 69:108277. [PMID: 37923235 DOI: 10.1016/j.biotechadv.2023.108277] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/07/2023]
Abstract
With the thrive of nanotechnology, silica nanoparticles (SiNPs) have been extensively adopted in the agriculture, food, cosmetic, and even biomedical industries. Due to the mass production and use, SiNPs inevitably entered the environment, resulting in ecological toxicity and even posing a threat to human health. Although considerable investigations have been conducted to assess the toxicity of SiNPs, the correlation between SiNPs exposure and consequent health risks remains ambiguous. Since the biological impacts of SiNPs can differ from their design and application, the toxicity assessment for SiNPs may be extremely difficult. This review discussed the application of SiNPs in different fields, especially their biomedical use, and documented their potential release pathways into the environment. Meanwhile, the current process of assessing SiNPs-related toxicity on various model organisms and cell lines was also detailed, thus estimating the health threats posed by SiNPs exposure. Finally, the potential toxic mechanisms of SiNPs were also elaborated based on results obtained from both in vivo and in vitro trials. This review generally summarizes the biological effects of SiNPs, which will build up a comprehensive perspective of the application and toxicity of SiNPs.
Collapse
Affiliation(s)
- Ruiyang Ding
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yang Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yang Yu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China.
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
12
|
Kausar S, Jabeen F, Latif MA, Asad M. Characterization, dose dependent assessment of hepatorenal oxidative stress, hematological parameters and histopathological divulging of the hepatic damages induced by Zinc oxide nanoparticles (ZnO-NPs) in adult male Sprague Dawley rats. Saudi J Biol Sci 2023; 30:103745. [PMID: 37588571 PMCID: PMC10425408 DOI: 10.1016/j.sjbs.2023.103745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/06/2023] [Accepted: 07/14/2023] [Indexed: 08/18/2023] Open
Abstract
Nanoparticles are beneficial in many aspects to human life but their excessive use can cause various abnormalities. They dispose in the environment through transport, industrial and agricultural usage and enter in living body through dermal, respiratory route or ingested with the lipsticks and there higher concentration produces toxicity. Therefore, current study characterized ZnO-NPs to evaluate toxic ability by X-rays diffraction (XRD) and Scanning Electron Microscopy (SEM) techniques and showed 29.83 and 35 nm size, respectively with hexagonal crystalline structure. LC50 value of ZnO-NPs was also evaluated as 72.48 ± 10.33 mg/kg BW. Male Sprague Dawley (Post weaning) rats were divided into five groups with five rats in each group. Control (C) group received no treatment, placebo (S) group received normal saline (0.9% sodium chloride) intraperitoneally and three treated groups received different levels of ZnO- NPs intraperitoneally at the dose of either 10 or 20 or 30 mg/kg for 21 days on alternate days and named as 1G1, 1G2 and 1G3, respectively for the assessment of toxicity for better understanding of precautionary measures in future. Oxidative stress enzymes of liver and kidney, hepatorenal function enzymes and hematological parameters along with hepatic histology were measured at the end of the experiment. Results showed highly significant variations in all parameters in a dose dependent manner as compared to control group while groups receiving 10 or 20 mg/kg of ZnO-NPs showed low to moderate pathological changes in both organs. Liver histological analysis showed congestion, necrosis, hemorrhage, RBC's accumulations; inflammatory cells infiltration and severe abnormalities in high dose group while medium, low dose group showed moderate and least effects, respectively. It is concluded that ZnO-NPs are highly toxic at more concentration so their careful usage is needed in daily routine.
Collapse
Affiliation(s)
- Sana Kausar
- Department of Zoology, Government College Universisty, Faisalabad, Pakistan
| | - Farhat Jabeen
- Department of Zoology, Government College Universisty, Faisalabad, Pakistan
| | | | - Muhammad Asad
- Department of Zoology, University of Education, Lahore, Pakistan
| |
Collapse
|
13
|
Xuan L, Ju Z, Skonieczna M, Zhou P, Huang R. Nanoparticles-induced potential toxicity on human health: Applications, toxicity mechanisms, and evaluation models. MedComm (Beijing) 2023; 4:e327. [PMID: 37457660 PMCID: PMC10349198 DOI: 10.1002/mco2.327] [Citation(s) in RCA: 129] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/04/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023] Open
Abstract
Nanoparticles (NPs) have become one of the most popular objects of scientific study during the past decades. However, despite wealth of study reports, still there is a gap, particularly in health toxicology studies, underlying mechanisms, and related evaluation models to deeply understanding the NPs risk effects. In this review, we first present a comprehensive landscape of the applications of NPs on health, especially addressing the role of NPs in medical diagnosis, therapy. Then, the toxicity of NPs on health systems is introduced. We describe in detail the effects of NPs on various systems, including respiratory, nervous, endocrine, immune, and reproductive systems, and the carcinogenicity of NPs. Furthermore, we unravels the underlying mechanisms of NPs including ROS accumulation, mitochondrial damage, inflammatory reaction, apoptosis, DNA damage, cell cycle, and epigenetic regulation. In addition, the classical study models such as cell lines and mice and the emerging models such as 3D organoids used for evaluating the toxicity or scientific study are both introduced. Overall, this review presents a critical summary and evaluation of the state of understanding of NPs, giving readers more better understanding of the NPs toxicology to remedy key gaps in knowledge and techniques.
Collapse
Affiliation(s)
- Lihui Xuan
- Department of Occupational and Environmental HealthXiangya School of Public HealthCentral South UniversityChangshaHunanChina
| | - Zhao Ju
- Department of Occupational and Environmental HealthXiangya School of Public HealthCentral South UniversityChangshaHunanChina
| | - Magdalena Skonieczna
- Department of Systems Biology and EngineeringInstitute of Automatic ControlSilesian University of TechnologyGliwicePoland
- Biotechnology Centre, Silesian University of TechnologyGliwicePoland
| | - Ping‐Kun Zhou
- Beijing Key Laboratory for RadiobiologyDepartment of Radiation BiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Ruixue Huang
- Department of Occupational and Environmental HealthXiangya School of Public HealthCentral South UniversityChangshaHunanChina
| |
Collapse
|
14
|
D'Iglio C, Famulari S, Capparucci F, Gervasi C, Cuzzocrea S, Spanò N, Di Paola D. Toxic Effects of Gemcitabine and Paclitaxel Combination: Chemotherapy Drugs Exposure in Zebrafish. TOXICS 2023; 11:544. [PMID: 37368644 DOI: 10.3390/toxics11060544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/09/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023]
Abstract
Pharmaceuticals are widely recognized as potentially hazardous to aquatic ecosystems. In the last two decades, the constant intake of biologically active chemicals used in human healthcare has been related to the growing release of these agents into natural environments. As reported by several studies, various pharmaceuticals have been detected, mainly in surface water (seas, lakes, and rivers), but also in groundwater and drinking water. Moreover, these contaminants and their metabolites can show biological activity even at very low concentrations. This study aimed to evaluate the developmental toxicity of exposure to the chemotherapy drugs gemcitabine and paclitaxel in aquatic environments. Zebrafish (Danio rerio) embryos were exposed to doses of gemcitabine 15 μM in combination with paclitaxel 1 μM from 0 to 96 h post-fertilization (hpf) using a fish embryo toxicity test (FET). This study highlights that both gemcitabine and paclitaxel exposure at single non-toxic concentrations affected survival and hatching rate, morphology score, and body length after exposure in combination. Additionally, exposure significantly disturbed the antioxidant defense system and increased ROS in zebrafish larvae. Gemcitabine and paclitaxel exposure caused changes in the expression of inflammation-related, endoplasmic reticulum stress-related (ERS), and autophagy-related genes. Taken together, our findings underline that gemcitabine and paclitaxel increase developmental toxicity in zebrafish embryos in a time-dependent manner.
Collapse
Affiliation(s)
- Claudio D'Iglio
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy
| | - Sergio Famulari
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy
| | - Fabiano Capparucci
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy
| | - Claudio Gervasi
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, 1402 S. Grand Blvd., St. Louis, MO 63104, USA
| | - Nunziacarla Spanò
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy
| | - Davide Di Paola
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, 98166 Messina, Italy
| |
Collapse
|
15
|
Gao X, Ma C, Wang H, Zhang C, Huang Y. Multi-walled carbon nanotube induced liver injuries possibly by promoting endoplasmic reticulum stress in Cyprinus carpio. CHEMOSPHERE 2023; 325:138383. [PMID: 36907489 DOI: 10.1016/j.chemosphere.2023.138383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 06/18/2023]
Abstract
The mass production and discharge of carbon nanotubes (CNTs) to the water environment are of great concern since they threaten the health of organisms in the aquatic ecosystem. CNTs induce multi-organ injuries in fish, but limited literature is available regarding the mechanisms involved. In the present study, juvenile common carp (Cyprinus carpio) were exposed to multi-walled carbon nanotubes (MWCNTs) (0.25 mg L-1 and 2.5 mg L-1) for four weeks. MWCNTs caused dose-dependent alterations in the pathological morphology of liver tissues. Ultrastructural changes manifested as nuclear deformation, chromatin condensation, endoplasmic reticulum (ER) disorderly arrangement, mitochondria vacuolation, and mitochondrial membrane destruction. TUNEL analysis indicated that the apoptosis rate in hepatocytes markedly increased upon exposure to MWCNTs. Moreover, the apoptosis was confirmed by significant upregulation of mRNA levels of apoptosis-related genes (Bcl-2, XBP1, Bax, and caspase3) in MWCNTs-exposure groups, except for Bcl-2 expression which was not significantly changed in HSC groups (2.5 mg L-1 MWCNTs). Furthermore, real-time PCR assay indicated the increased expression of ER stress (ERS) marker genes (GRP78, PERK, and eIF2α) in the exposure groups compared to the control groups, suggesting that the PERK/eIF2α signaling pathway involved in the injuries of the liver tissue. Overall, the results above indicate that MWCNTs induce ERS by activating the PERK/eIF2α pathway in the liver of common carp, and resulted in the initiation of apoptosis procedure.
Collapse
Affiliation(s)
- Xiaochan Gao
- School of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471003, China.
| | - Chaoran Ma
- School of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471003, China.
| | - Hongjun Wang
- School of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471003, China.
| | - Chunnuan Zhang
- School of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471003, China.
| | - Yong Huang
- School of Animal Science and Technology, Henan University of Science and Technology, Luoyang, 471003, China.
| |
Collapse
|
16
|
Lv R, Liu X, Zhang Y, Dong N, Wang X, He Y, Yue H, Yin Q. Pathophysiological mechanisms and therapeutic approaches in obstructive sleep apnea syndrome. Signal Transduct Target Ther 2023; 8:218. [PMID: 37230968 DOI: 10.1038/s41392-023-01496-3] [Citation(s) in RCA: 136] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023] Open
Abstract
Obstructive sleep apnea syndrome (OSAS) is a common breathing disorder in sleep in which the airways narrow or collapse during sleep, causing obstructive sleep apnea. The prevalence of OSAS continues to rise worldwide, particularly in middle-aged and elderly individuals. The mechanism of upper airway collapse is incompletely understood but is associated with several factors, including obesity, craniofacial changes, altered muscle function in the upper airway, pharyngeal neuropathy, and fluid shifts to the neck. The main characteristics of OSAS are recurrent pauses in respiration, which lead to intermittent hypoxia (IH) and hypercapnia, accompanied by blood oxygen desaturation and arousal during sleep, which sharply increases the risk of several diseases. This paper first briefly describes the epidemiology, incidence, and pathophysiological mechanisms of OSAS. Next, the alterations in relevant signaling pathways induced by IH are systematically reviewed and discussed. For example, IH can induce gut microbiota (GM) dysbiosis, impair the intestinal barrier, and alter intestinal metabolites. These mechanisms ultimately lead to secondary oxidative stress, systemic inflammation, and sympathetic activation. We then summarize the effects of IH on disease pathogenesis, including cardiocerebrovascular disorders, neurological disorders, metabolic diseases, cancer, reproductive disorders, and COVID-19. Finally, different therapeutic strategies for OSAS caused by different causes are proposed. Multidisciplinary approaches and shared decision-making are necessary for the successful treatment of OSAS in the future, but more randomized controlled trials are needed for further evaluation to define what treatments are best for specific OSAS patients.
Collapse
Affiliation(s)
- Renjun Lv
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Xueying Liu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Yue Zhang
- Department of Geriatrics, the 2nd Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Na Dong
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Xiao Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Yao He
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Hongmei Yue
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
| | - Qingqing Yin
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.
| |
Collapse
|
17
|
Yang C, Lu Z, Xia Y, Zhang J, Zou Z, Chen C, Wang X, Tian X, Cheng S, Jiang X. Alterations of Gut-Derived Melatonin in Neurobehavioral Impairments Caused by Zinc Oxide Nanoparticles. Int J Nanomedicine 2023; 18:1899-1914. [PMID: 37057188 PMCID: PMC10088905 DOI: 10.2147/ijn.s386240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 12/24/2022] [Indexed: 04/15/2023] Open
Abstract
Purpose The widespread use of zinc oxide nanoparticles (ZnONPs) has raised concerns about its potential toxicity. Melatonin is a neurohormone with tremendous anti-toxic effects. The enterochromaffin cells are an essential source of melatonin in vivo. However, studies on the effects of ZnONPs on endogenous melatonin are minimal. In the present study, we aimed to investigate the effects of ZnONPs exposure on gut-derived melatonin. Methods In the present study, 64 adult male mice were randomly and equally divided into four groups, and each group was exposed to ZnONPs (0, 6.5, 13, 26 mg/kg/day) for 30 days. Subsequently, the neurobehavioral changes were observed. The effects of ZnONPs on the expression of melatonin-related genes arylalkylamine N-acetyltransferase (Aanat), melatonin receptor1A (Mt1/Mtnr1a), melatonin receptor1B (Mt2/Mtnr1b), and neuropeptide Y (Npy) on melatonin synthesis and secretion in duodenum, jejunum, ileum and colon during day and night were also assessed. Results The results revealed that oral exposure to ZnONPs induced impairments of locomotor activity and anxiety-like behavior in adult mice during the day. The transcriptional analysis of brain tissues revealed that exposure to ZnONPs caused profound effects on genes and transcriptional signaling pathways associated with melatonin synthesis and metabolic processes during the day and night. We also observed that, in the duodenum, jejunum, ileum and colon sites, ZnONPs resulted in a significant reduction in the expression of the gut-derived melatonin rate-limiting enzyme Aanat, the membrane receptors Mt1 and Mt2 and Npy during the day and night. Conclusion Taken together, this is the first study shows that oral exposure to ZnONPs interferes with melatonin synthesis and secretion in different intestinal segments of adult mice. These findings will provide novelty insights into the neurotoxic mechanisms of ZnONPs and suggest an alternative strategy for the prevention of ZnONP neurotoxicity.
Collapse
Affiliation(s)
- Cantao Yang
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Zhaohong Lu
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yinyin Xia
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Jun Zhang
- Molecular Biology Laboratory of Respiratory Diseases, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Zhen Zou
- Molecular Biology Laboratory of Respiratory Diseases, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Xiaoliang Wang
- Medical Sciences Research Center, University-Town Hospital of Chongqing Medical University, Chongqing, 401331, People’s Republic of China
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Shuqun Cheng
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Correspondence: Shuqun Cheng; Xuejun Jiang, Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Number 1, Yixueyuan Road, Yuzhong District, Chongqing, 400016, People’s Republic of China, Tel +86-23-68485008, Fax +86-23-68485207, Email ;
| | - Xuejun Jiang
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
- Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| |
Collapse
|
18
|
Biocidal activity of ZnO NPs against pathogens and antioxidant activity - a greener approach by Citrus hystrix leaf extract as bio-reductant. Biochem Eng J 2023. [DOI: 10.1016/j.bej.2023.108818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
19
|
Role of ER Stress in Xenobiotic-Induced Liver Diseases and Hepatotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4640161. [PMID: 36388166 PMCID: PMC9652065 DOI: 10.1155/2022/4640161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/12/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
Abstract
The liver is a highly metabolic organ and plays a crucial role in the transportation, storage, and/or detoxication of xenobiotics. Liver damage induced by xenobiotics (e.g., heavy metal, endocrine disrupting chemicals, Chinese herbal medicine, or nanoparticles) has become a pivotal reason for liver diseases, leading to great clinical challenge and much attention for the past decades. Given that endoplasmic reticulum (ER) is the prominent organelle involved in hepatic metabolism, ER dysfunction, namely, ER stress, is clearly observed in various liver diseases. In response to ER stress, a conserved adaptive signaling pathway known as unfolded protein response (UPR) is activated to restore ER homeostasis. However, the prolonged ER stress with UPR eventually leads to the death of hepatocytes, which is a pathogenic event in many hepatic diseases. Therefore, analyzing the perturbation in the activation or inhibition of ER stress and the UPR signaling pathway is likely an effective marker for investigating the molecular mechanisms behind the toxic effects of xenobiotics on the liver. We review the role of ER stress in hepatic diseases and xenobiotic-induced hepatotoxicity, which not only provides a theoretical basis for further understanding the pathogenesis of liver diseases and the mechanisms of hepatotoxicity induced by xenobiotics but also presents a potential target for the prevention and treatment of xenobiotic-related liver diseases.
Collapse
|
20
|
Zearalenone Promotes LPS-Induced Oxidative Stress, Endoplasmic Reticulum Stress, and Accelerates Bovine Mammary Epithelial Cell Apoptosis. Int J Mol Sci 2022; 23:ijms231810925. [PMID: 36142835 PMCID: PMC9500836 DOI: 10.3390/ijms231810925] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/05/2022] [Accepted: 09/13/2022] [Indexed: 11/29/2022] Open
Abstract
Both zearalenone (ZEA) and lipopolysaccharide (LPS) can induce oxidative stress, and even apoptosis in bovine mammary epithelial cells (MAC-T), but not much attention has been given to the synergistic effect of ZEA and LPS. In this study, we treated MAC-T cells with different concentrations of LPS (1, 10, 50, and 100 μg/mL) and ZEA (5, 15, and 30 μM) to induce cell damage. Previous results show that MAC-T cell viability decreases with increasing LPS concentration. Meanwhile, 1 µg/mL LPS and ZEA were selected for combined treatment in subsequent studies. It was found that co-treatment with ZEA and LPS increases the accumulation of reactive oxygen species (ROS) and malondialdehyde (MDA), decreases mitochondrial membrane potential (MMP), and superoxide dismutase (SOD), and reduces glutathione (GSH). ZEA and LPS are found to activate endoplasmic reticulum (ER) stress by increasing the expression of glucose-regulated protein 78 kDa (GRP78), activating transcription factor 6 (ATF6) and C/EBP homologous protein (CHOP). It increases cell apoptosis by suppressing the expression of the anti-apoptotic protein B-cell lymphoma-2 (Bcl-2), indicated by up-regulation of Bcl2-associated X protein (Bax) and Cysteinyl aspartate-specific proteinases 3 (caspase-3) expression. The above results suggest that the synergistic effect of ZEA and LPS aggravate cytotoxicity.
Collapse
|
21
|
Cameron SJ, Sheng J, Hosseinian F, Willmore WG. Nanoparticle Effects on Stress Response Pathways and Nanoparticle-Protein Interactions. Int J Mol Sci 2022; 23:7962. [PMID: 35887304 PMCID: PMC9323783 DOI: 10.3390/ijms23147962] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/01/2022] [Accepted: 07/11/2022] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles (NPs) are increasingly used in a wide variety of applications and products; however, NPs may affect stress response pathways and interact with proteins in biological systems. This review article will provide an overview of the beneficial and detrimental effects of NPs on stress response pathways with a focus on NP-protein interactions. Depending upon the particular NP, experimental model system, and dose and exposure conditions, the introduction of NPs may have either positive or negative effects. Cellular processes such as the development of oxidative stress, the initiation of the inflammatory response, mitochondrial function, detoxification, and alterations to signaling pathways are all affected by the introduction of NPs. In terms of tissue-specific effects, the local microenvironment can have a profound effect on whether an NP is beneficial or harmful to cells. Interactions of NPs with metal-binding proteins (zinc, copper, iron and calcium) affect both their structure and function. This review will provide insights into the current knowledge of protein-based nanotoxicology and closely examines the targets of specific NPs.
Collapse
Affiliation(s)
- Shana J. Cameron
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
| | - Jessica Sheng
- Department of Biology, Carleton University, Ottawa, ON K1S 5B6, Canada;
| | - Farah Hosseinian
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
| | - William G. Willmore
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
- Department of Biology, Carleton University, Ottawa, ON K1S 5B6, Canada;
- Institute of Biochemistry, Carleton University, Ottawa, ON K1S 5B6, Canada
| |
Collapse
|
22
|
Lei R, Zhou M, Zhang S, Luo J, Qu C, Wang Y, Guo P, Huang R. Potential role of PRKCSH in lung cancer: bioinformatics analysis and a case study of Nano ZnO. NANOSCALE 2022; 14:4495-4510. [PMID: 35254362 DOI: 10.1039/d1nr08133k] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
PRKCSH, also known as glucosidase II beta, functions as a contributor to lung tumorigenesis by regulating the cell cycle in a p53-dependent manner under severe environmental stress. However, the prognostic value and molecular mechanisms by which the level of PRKCSH is significantly increased in cancer cells are not clearly understood. Here, we first generated a biological profile of PRKCSH expression changes in cancers by analysing bioinformatic data from cancer databases. We found that higher PRKCSH expression was correlated with a poorer prognosis and greater infiltration of most immune cell types in patients with lung cancer. In particular, PRKCSH expression showed significant negative correlations with the level of STAT6 (r = -0.31, p < 0.001) in lung cancer tissues. We further found that PRKCSH deficiency promoted G2/M arrest in response to zinc oxide nanoparticle (Nano ZnO) treatment in A549 cells. With regard to the mechanism, PRKCSH deficiency may induce STAT6 translocation to the nucleus to activate p53 expression through binding to the p53 promoter region from -365 bp to +126 bp. Eventually, activated p53 contributed to Nano-ZnO-induced G2/M arrest in lung cancer cells. Taken together, our data provide new insights into immunotherapy target choices and the prognostic value of PRKCSH. Since the G2/M cell cycle checkpoint is crucial for lung cancer prognosis, targeting PRKCSH expression to suppress the activation of the STAT6/p53 pathway is a potential therapeutic strategy for managing lung cancer.
Collapse
Affiliation(s)
- Ridan Lei
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China.
| | - Meiling Zhou
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China.
| | - Shusheng Zhang
- Changsha Stomatological Hospital, Changsha, Hunan Province, China.
| | - Jinhua Luo
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China.
| | - Can Qu
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China.
| | - Yin Wang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China.
| | - Peiyu Guo
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China.
| | - Ruixue Huang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan Province 410078, China.
| |
Collapse
|
23
|
Double-Sided Nano-ZnO: Superior Antibacterial Properties and Induced Hepatotoxicity in Zebrafish Embryos. TOXICS 2022; 10:toxics10030144. [PMID: 35324769 PMCID: PMC8950655 DOI: 10.3390/toxics10030144] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/07/2022] [Accepted: 03/16/2022] [Indexed: 12/30/2022]
Abstract
Zinc oxide nanoparticles (Nano-ZnO) have been widely used in the food, cosmetics, and biomedical fields due to their excellent antibacterial and antioxidant properties. However, with the widespread application of Nano-ZnO, Nano-ZnO inevitably enters the environment and living organisms, causing harm to human health and ecosystem safety. Therefore, the biosafety and toxicological issues of Nano-ZnO are gradually being emphasized. Our study found that Nano-ZnO has superior antibacterial properties compared to ofloxacin in the fight against Staphylococcus aureus (S. aureus). Given that ofloxacin can inhibit bacterial-induced inflammation, we constructed a model of bacterial inflammation using S. aureus in zebrafish. We found that Nano-ZnO inhibited the NF-κB-mediated inflammatory signaling pathway. However, in the process, we found that Nano-ZnO caused hepatic steatosis in zebrafish. This suggested that Nano-ZnO had a certain hepatotoxicity, but did not affect liver development. Subsequently, we investigated the mechanism of hepatotoxicity produced by Nano-ZnO. Nano-ZnO triggered oxidative stress in the liver by generating ROS, which then induced endoplasmic reticulum stress to occur. It further activated srebp and its downstream genes fasn and acc1, which promoted the accumulation of fatty acid synthesis and the development of steatosis, leading to the development of nonalcoholic fatty liver disease (NAFLD). To address the hepatotoxicity of Nano-ZnO, we added carbon dots for the treatment of NAFLD. The carbon dots were found to normalize the steatotic liver. This provided a new strategy to address the hepatotoxicity caused by Nano-ZnO. In this work, we systematically analyzed the antibacterial advantages of Nano-ZnO in vivo and in vitro, explored the mechanism of Nano-ZnO hepatotoxicity, and proposed a new method to treat Nano-ZnO hepatotoxicity.
Collapse
|
24
|
Warsi AZ, Aziz F, Zulfiqar S, Haider S, Shakir I, Agboola PO. Synthesis, Characterization, Photocatalysis, and Antibacterial Study of WO 3, MXene and WO 3/MXene Nanocomposite. NANOMATERIALS 2022; 12:nano12040713. [PMID: 35215041 PMCID: PMC8877483 DOI: 10.3390/nano12040713] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 02/07/2023]
Abstract
Tungsten oxide (WO3), MXene, and an WO3/MXene nanocomposite were synthesized to study their photocatalytic and biological applications. Tungsten oxide was synthesized by an easy and cost-effective hydrothermal method, and its composite with MXene was prepared through the sonication method. The synthesized tungsten oxide, MXene, and its composite were characterized by X-ray diffraction (XRD), field emission scanning electron microscopy (FESEM), Fourier transform infrared (FTIR), energy-dispersive X-ray analysis (EDX), and Brunauer–Emmett–Teller (BET) for their structural, morphological, spectral, elemental and surface area analysis, respectively. The crystallite size of WO3 calculated from XRD was ~10 nm, the particle size of WO3 was 130 nm, and the average thickness of MXene layers was 175 nm, which was calculated from FESEM. The photocatalytic activity of as-synthesized samples was carried out for the degradation of methylene blue under solar radiation, MXene, the WO3/MXene composite, and WO3 exhibited 54%, 89%, and 99% photocatalytic degradation, respectively. WO3 showed maximal degradation ability; by adding WO3 to MXene, the degradation ability of MXene was enhanced. Studies on antibacterial activity demonstrated that these samples are good antibacterial agents against positive strains, and their antibacterial activity against negative strains depends upon their concentration. Against positive strains, the WO3/MXene composite’s inhibition zone was at 7 mm, while it became 9 mm upon increasing the concentration. This study proves that WO3, MXene, and the WO3/MXene nanocomposite could be used in biological and environmental applications.
Collapse
Affiliation(s)
- Al-Zoha Warsi
- Baghdad-ul-Jadeed Campus, Institute of Chemistry, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan; (A.-Z.W.); (F.A.)
| | - Fatima Aziz
- Baghdad-ul-Jadeed Campus, Institute of Chemistry, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan; (A.-Z.W.); (F.A.)
| | - Sonia Zulfiqar
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo, New Cairo 11835, Egypt;
| | - Sajjad Haider
- Department of Chemical Engineering, College of Engineering, King Saud University, P.O. Box 800, Riyadh 11421, Saudi Arabia;
| | - Imran Shakir
- Sustainable Energy Technologies (SET) Center, College of Engineering, King Saud University, P.O. Box 800, Riyadh 11421, Saudi Arabia;
| | - Philips O. Agboola
- College of Engineering, Al-Muzahmia Branch, King Saud University, P.O. Box 800, Riyadh 11421, Saudi Arabia
- Correspondence:
| |
Collapse
|
25
|
Zhang Y, Liu B, Liu Z, Li J. Research progress in synthesis and biological application of quantum dots. NEW J CHEM 2022. [DOI: 10.1039/d2nj02603a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Quantum dots are an excellent choice for biomedical applications due to their special optical properties and quantum confinement effects. This paper reviews the research and application progress of several quantum...
Collapse
|
26
|
Hu H, Luo Z, Liu X, Huang L, Lu X, Ding R, Duan C, He Y. Sestrin2 Overexpression Ameliorates Endoplasmic Reticulum Stress-Induced Apoptosis via Inhibiting mTOR Pathway in HepG2 Cells. Int J Endocrinol 2022; 2022:2009753. [PMID: 36536875 PMCID: PMC9759384 DOI: 10.1155/2022/2009753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/08/2022] [Accepted: 11/21/2022] [Indexed: 12/14/2022] Open
Abstract
Sestrin2 is a highly conserved stress-inducible protein, acting as a crucial part in regulating homeostasis in response to various stress conditions in the cell. However, the role of Sestrin2 in regulating cell apoptosis related to endoplasmic reticulum (ER) has not been fully investigated. Our study presented here aims to reveal the effect of Sestrin2 in tunicamycin (TM)-induced cell apoptosis related to ER stress and its underlying molecular mechanisms. The results demonstrated that Sestrin2 expression was significantly upregulated correlated with ER stress responses in TM treated HepG2 cells. Sestrin2 overexpression obviously alleviated ER stress with the determination of ER stress-related proteins expression. In addition, Sestrin2 overexpression inhibited cell apoptosis with the examination of apoptosis-related proteins and TUNEL assay. However, Sestrin2 knockdown further promoted the ER stress-mediated cell apoptosis. The further mechanistic study revealed that Sestrin2 overexpression inhibited TM-induced mTOR pathway activation. Taken together, our current study indicated that Sestrin2 overexpression ameliorates ER stress-induced apoptosis via inhibiting mTOR pathway in HepG2 cells.
Collapse
Affiliation(s)
- Huiling Hu
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhijun Luo
- Department of Emergency, The Seventh Affiliated Hospital, Southern Medical University, Foshan, Guangdong 528244, China
| | - Xiuli Liu
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lisi Huang
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaoxia Lu
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Rui Ding
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chaohui Duan
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yuqing He
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
27
|
Dutta G, Sugumaran A. Bioengineered zinc oxide nanoparticles: Chemical, green, biological fabrication methods and its potential biomedical applications. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102853] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
28
|
Mihailovic V, Katanic Stankovic JS, Selakovic D, Rosic G. An Overview of the Beneficial Role of Antioxidants in the Treatment of Nanoparticle-Induced Toxicities. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7244677. [PMID: 34820054 PMCID: PMC8608524 DOI: 10.1155/2021/7244677] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/26/2021] [Indexed: 12/20/2022]
Abstract
Nanoparticles (NPs) are used in many products and materials for humans such as electronics, in medicine for drug delivery, as biosensors, in biotechnology, and in agriculture, as ingredients in cosmetics and food supplements. Besides that, NPs may display potentially hazardous properties on human health and the environment as a consequence of their abundant use in life nowadays. Hence, there is increased interest of researchers to provide possible therapeutic agents or dietary supplements for the amelioration of NP-induced toxicity. This review summarizes the new findings in the research of the use of antioxidants as supplements for the prevention and alleviation of harmful effects caused by exposure of organisms to NPs. Also, mechanisms involved in the formation of NP-induced oxidative stress and protective mechanisms using different antioxidant substances have also been elaborated. This review also highlights the potential of naturally occurring antioxidants for the enhancement of the antioxidant defense systems in the prevention and mitigation of organism damage caused by NP-induced oxidative stress. Based on the presented results of the most recent studies, it may be concluded that the role of antioxidants in the prevention and treatment of nanoparticle-induced toxicity is unimpeachable. This is particularly important in terms of oxidative stress suppression.
Collapse
Affiliation(s)
- Vladimir Mihailovic
- University of Kragujevac, Faculty of Science, Department of Chemistry, Radoja Domanovica 12, 34000 Kragujevac, Serbia
| | - Jelena S. Katanic Stankovic
- University of Kragujevac, Institute for Information Technologies Kragujevac, Department of Science, Jovana Cvijica bb, 34000 Kragujevac, Serbia
| | - Dragica Selakovic
- University of Kragujevac, Faculty of Medical Sciences, Department of Physiology, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Gvozden Rosic
- University of Kragujevac, Faculty of Medical Sciences, Department of Physiology, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| |
Collapse
|
29
|
Mihailovic V, Katanic Stankovic JS, Selakovic D, Rosic G. An Overview of the Beneficial Role of Antioxidants in the Treatment of Nanoparticle-Induced Toxicities. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021. [DOI: https://doi.org/10.1155/2021/7244677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Nanoparticles (NPs) are used in many products and materials for humans such as electronics, in medicine for drug delivery, as biosensors, in biotechnology, and in agriculture, as ingredients in cosmetics and food supplements. Besides that, NPs may display potentially hazardous properties on human health and the environment as a consequence of their abundant use in life nowadays. Hence, there is increased interest of researchers to provide possible therapeutic agents or dietary supplements for the amelioration of NP-induced toxicity. This review summarizes the new findings in the research of the use of antioxidants as supplements for the prevention and alleviation of harmful effects caused by exposure of organisms to NPs. Also, mechanisms involved in the formation of NP-induced oxidative stress and protective mechanisms using different antioxidant substances have also been elaborated. This review also highlights the potential of naturally occurring antioxidants for the enhancement of the antioxidant defense systems in the prevention and mitigation of organism damage caused by NP-induced oxidative stress. Based on the presented results of the most recent studies, it may be concluded that the role of antioxidants in the prevention and treatment of nanoparticle-induced toxicity is unimpeachable. This is particularly important in terms of oxidative stress suppression.
Collapse
Affiliation(s)
- Vladimir Mihailovic
- University of Kragujevac, Faculty of Science, Department of Chemistry, Radoja Domanovica 12, 34000 Kragujevac, Serbia
| | - Jelena S. Katanic Stankovic
- University of Kragujevac, Institute for Information Technologies Kragujevac, Department of Science, Jovana Cvijica bb, 34000 Kragujevac, Serbia
| | - Dragica Selakovic
- University of Kragujevac, Faculty of Medical Sciences, Department of Physiology, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Gvozden Rosic
- University of Kragujevac, Faculty of Medical Sciences, Department of Physiology, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| |
Collapse
|
30
|
Beladiya JV, Mehta AA. Acute and 28-days subacute toxicity studies of Gαq-RGS2 signaling inhibitor. Lab Anim Res 2021; 37:17. [PMID: 34311782 PMCID: PMC8314442 DOI: 10.1186/s42826-021-00093-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/26/2021] [Indexed: 11/25/2022] Open
Abstract
Background The aim of study was to evaluate the single oral dose and 28 day repeated oral administration toxicity profile of the synthetic compound Gαq-RGS2 signaling inhibitor, (1-(5-chloro-2-hydroxyphenyl)-3-(4-(trifluoromethyl)phenyl)-1 H-1,2,4-triazol-5(4 H)-one) as per OECD guideline 425 (2008a) and 407 (2008b), respectively. Results In acute toxicity study, a single oral dose administration of Gαq-RGS2 signaling inhibitor did not show any mortality at doses of 5, 50, 300 and 2000 mg/kg within 24 h and 14 days. The treatment of Gαq-RGS2 signaling inhibitor at dose 10 and 100 mg/kg for 28 days did not show any mortality, significant changes in the increase of body weight, various organ damage markers, hematological parameters, relative organ/body weight ratio and microscopic anatomical texture of essential organs as compared to vehicle and normal control. Conclusions A single oral administration of Gαq-RGS2 signaling inhibitor up to dose of 2000 mg/kg in mice and repeated administration of Gαq-RGS2 signaling inhibitor at higher dose 100 mg/kg for 28 days in the rats is safe. Supplementary Information The online version contains supplementary material available at 10.1186/s42826-021-00093-1.
Collapse
Affiliation(s)
- Jayesh V Beladiya
- Department of Pharmacology, L. M. College of Pharmacy, Navarangpura, Gujarat, 380009, Ahmedabad, India
| | - Anita A Mehta
- Department of Pharmacology, L. M. College of Pharmacy, Navarangpura, Gujarat, 380009, Ahmedabad, India.
| |
Collapse
|
31
|
Yang Y, Xia R, Zhang X, Wang X, Zhou Y, Wang H, Feng Y, Lv S, Ji S. Effects of Oral Exposure to Mn-Doped ZnS Quantum Dots on Intestinal Tract and Gut Microbiota in Mice. Front Physiol 2021; 12:657266. [PMID: 34295256 PMCID: PMC8290145 DOI: 10.3389/fphys.2021.657266] [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] [Received: 01/26/2021] [Accepted: 05/27/2021] [Indexed: 11/14/2022] Open
Abstract
Mn-doped ZnS quantum dots (QDs) with excellent optical properties have been explored in a wide range of fields. Their potential adverse effects on biological systems and human health should be evaluated before biological application. In the present study, we investigated the effect of Mn-doped ZnS QDs on the intestinal tract and gut microbiota structures at 2 h and 14 days (d) after 14 d repeated oral exposure in mice. Flame atomic absorption spectrophotometry (FAAS), histopathological examination, and transmission electron microscopy (TEM) were used to assess the absorption and toxicity of Mn-doped ZnS QDs on the intestinal tract. The 16S rRNA gene sequencing was used to evaluate the gut microbial communities. Mn-doped ZnS QDs did not accumulate in the duodenum, jejunum, ileum, or colon. The Zn content of feces was not significantly higher than in the control group. No major histological changes were found in these tissues. The intestinal microvilli remained regular, but swelling of mitochondria and endoplasmic reticulum was detected by TEM at 14 d after the last gavage. A total of 2,712 operational taxonomic units (OTUs) were generated. Mn-doped ZnS QDs treatment did not significantly change the α-diversity of Richness, Chao1, Shannon, and Simpson indexes. According to principal component analysis (PCA), Mn-doped ZnS QDs had no effect on the overall structure of the gut microbiota. No significant change occurred at the phylum level, while three genera were downregulated at 2 h and seven changed at 14 d after the last gavage. Our findings revealed that Mn-doped ZnS QDs had a little stimulation of the intestinal tract and gut microbiota, and oral administration may be a safe route for biological application (such as bioimaging and drug delivery).
Collapse
Affiliation(s)
- Yanjie Yang
- Institute of Molecular Medicine, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Ruixue Xia
- Institute of Molecular Medicine, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Xiaomei Zhang
- Institute of Molecular Medicine, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Xu Wang
- Institute of Molecular Medicine, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yuchen Zhou
- Institute of Molecular Medicine, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Honggang Wang
- Institute of Molecular Medicine, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yu Feng
- Institute of Molecular Medicine, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Shuangyu Lv
- Institute of Molecular Medicine, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Shaoping Ji
- Institute of Molecular Medicine, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| |
Collapse
|
32
|
Chang X, Tian M, Zhang Q, Liu F, Gao J, Li S, Liu H, Hou X, Li L, Li C, Sun Y. Grape seed proanthocyanidin extract ameliorates cisplatin-induced testicular apoptosis via PI3K/Akt/mTOR and endoplasmic reticulum stress pathways in rats. J Food Biochem 2021; 45:e13825. [PMID: 34152018 DOI: 10.1111/jfbc.13825] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/29/2021] [Accepted: 05/29/2021] [Indexed: 12/14/2022]
Abstract
Testicular toxicity is an adverse reaction of the effective chemotherapy drug cisplatin (CIS). Our previous study found that grape seed proanthocyanidin extract (GSPE) had a protective effect on CIS-induced testicular toxicity. However, the protective mechanism of GSPE against CIS-induced testicular toxicity remains unknown. In this study, we aimed to investigate whether GSPE can reduce CIS-induced testicular toxicity and its potential mechanism in rats. The results showed that GSPE ameliorated CIS-induced the apoptosis of testicular cells and inhibited the protein levels of Bad, Cyt c, caspase-9, caspase-3, caspase-12, GRP78, CHOP, IRE1α, p-IRE1α, XBP-1S, PERK, p-PERK, eIF2α, and p-eIF2α. Besides, GSPE reversed the downregulation of PI3K, p-PI3K, Akt, p-Akt, mTOR, and p-mTOR protein expression induced by CIS. These results indicated that GSPE can improve CIS-induced testicular cells apoptosis via activating PI3K/Akt/mTOR and inhibiting Bad/Cyt c/caspase-9/caspase-3 pathways. And GSPE relieved endoplasmic reticulum stress-mediated apoptosis via inhibiting PREK/eIF2α and IRE1α/XBP-1S/caspase-12 pathways. In conclusion, the evidence suggested that GSPE can act as a protective agent against testicular toxicity induced by CIS. PRACTICAL APPLICATIONS: Testicular toxicity was a well-known adverse effect of cisplatin (CIS) in cancer treatment. Grape seed proanthocyanidin extract (GSPE) has been reported to serve as one of the most therapeutic potentials agents. In present study, we explored the regulatory effects of GSPE on the apoptosis induced by CIS, which involved testicular apoptosis mechanisms in rats. Our results indicated that CIS caused testicular toxicity via PI3K/AKT/mTOR and ERS mediated apoptosis pathway in rats. This toxicity was attenuated by GSPE treatment via activated PI3K/Akt/mTOR pathway, and inhibiting Bad/CytC/caspase-9/caspase-3 as well as PREK/eIF2α, IRE1α/XBP-1S/caspase-12 pathways. Our findings suggest that GSPE may be a novel protective agent against testicular toxicity induced by CIS.
Collapse
Affiliation(s)
- Xuhong Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Minmin Tian
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Qiong Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Fangfang Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Jinxia Gao
- Department of Occupational Diseases, Lanzhou Municipal Center for Disease Control, Lanzhou, China
| | - Sheng Li
- Department of Public Health, The First People's Hospital of Lanzhou City, Lanzhou, China
| | - Han Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Xiangbo Hou
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Lei Li
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Chengyun Li
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Yingbiao Sun
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| |
Collapse
|
33
|
Gurunathan S, Kang MH, Jeyaraj M, Kim JH. Palladium Nanoparticle-Induced Oxidative Stress, Endoplasmic Reticulum Stress, Apoptosis, and Immunomodulation Enhance the Biogenesis and Release of Exosome in Human Leukemia Monocytic Cells (THP-1). Int J Nanomedicine 2021; 16:2849-2877. [PMID: 33883895 PMCID: PMC8055296 DOI: 10.2147/ijn.s305269] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/11/2021] [Indexed: 12/23/2022] Open
Abstract
Background Exosomes are endosome-derived nano-sized vesicles that have emerged as important mediators of intercellular communication and play significant roles in various diseases. However, their applications are rigorously restricted by the limited secretion competence of cells. Therefore, strategies to enhance the production and functions of exosomes are warranted. Studies have shown that nanomaterials can significantly enhance the effects of cells and exosomes in intercellular communication; however, how palladium nanoparticles (PdNPs) enhance exosome release in human leukemia monocytic cells (THP-1) remains unclear. Therefore, this study aimed to address the effect of PdNPs on exosome biogenesis and release in THP-1 cells. Methods Exosomes were isolated by ultracentrifugation and ExoQuickTM and characterized by dynamic light scattering, nanoparticle tracking analysis system, scanning electron microscopy, transmission electron microscopy, EXOCETTM assay, and fluorescence polarization. The expression levels of exosome markers were analyzed via quantitative reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay. Results PdNP treatment enhanced the biogenesis and release of exosomes by inducing oxidative stress, endoplasmic reticulum stress, apoptosis, and immunomodulation. The exosomes were spherical in shape and had an average diameter of 50–80 nm. Exosome production was confirmed via total protein concentration, exosome counts, acetylcholinesterase activity, and neutral sphingomyelinase activity. The expression levels of TSG101, CD9, CD63, and CD81 were significantly higher in PdNP-treated cells than in control cells. Further, cytokine and chemokine levels were significantly higher in exosomes isolated from PdNP-treated THP-1 cells than in those isolated from control cells. THP-1 cells pre-treated with N-acetylcysteine or GW4869 showed significant decreases in PdNP-induced exosome biogenesis and release. Conclusion To our knowledge, this is the first study showing that PdNPs stimulate exosome biogenesis and release and simultaneously increase the levels of cytokines and chemokines by modulating various physiological processes. Our findings suggest a reasonable approach to improve the production of exosomes for various therapeutic applications.
Collapse
Affiliation(s)
| | - Min-Hee Kang
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, Korea
| | - Muniyandi Jeyaraj
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, Korea
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, Korea
| |
Collapse
|
34
|
Liu L, Kong L. Research progress on the carcinogenicity of metal nanomaterials. J Appl Toxicol 2021; 41:1334-1344. [PMID: 33527484 DOI: 10.1002/jat.4145] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/16/2021] [Accepted: 01/22/2021] [Indexed: 12/21/2022]
Abstract
With the rapid development of nanotechnology, new nanomaterials with enormous potentials continue to emerge, especially metal nanomaterials. Metal nanomaterials possess the characteristics of metals and nanomaterials, so they are widely used in many fields. But at the same time, whether the use or release of metal nan4omaterials into the environment is toxic to human beings and animals has now attained widespread attention at home and abroad. Currently, it is an indisputable fact that cancer ranks among the top causes of death among residents worldwide. The properties of causing DNA damage and mutations possessed by these metal nanomaterials make them unpredictable influences in the body, subsequently leading to genotoxicity and carcinogenicity. Due to the increasing evidence of their roles in carcinogenicity, this article reviews the toxicological and carcinogenic effects of metal nanomaterials, including nano-metal elements (nickel nanoparticles, silver nanoparticles, and cobalt nanoparticles) and nano-metal oxides (titanium dioxide nanoparticles, silica nanoparticles, zinc oxide nanoparticles, and alumina nanoparticles). This article provides a reference for the researchers and policymakers to use metal nanomaterials rationally in modern industries and biomedicine.
Collapse
Affiliation(s)
- Lin Liu
- Key Laboratory of Environment Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Lu Kong
- Key Laboratory of Environment Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| |
Collapse
|
35
|
Ma RH, Ni ZJ, Thakur K, Zhang F, Zhang YY, Zhang JG, Wei ZJ. Natural Compounds Play Therapeutic Roles in Various Human Pathologies via Regulating Endoplasmic Reticulum Pathway. MEDICINE IN DRUG DISCOVERY 2020. [DOI: 10.1016/j.medidd.2020.100065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
36
|
Singh TA, Das J, Sil PC. Zinc oxide nanoparticles: A comprehensive review on its synthesis, anticancer and drug delivery applications as well as health risks. Adv Colloid Interface Sci 2020; 286:102317. [PMID: 33212389 DOI: 10.1016/j.cis.2020.102317] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 11/06/2020] [Accepted: 11/06/2020] [Indexed: 12/29/2022]
Abstract
In recent years, zinc oxide nanoparticles (ZnONPs) emerged as an excellent candidate in the field of optical, electrical, food packaging and particularly in biomedical research. ZnONPs show cancer cell specific toxicity via the pH-dependent (low pH) dissolution into Zn2+ ions, which generate reactive oxygen species and induce cytotoxicity in cancer cells. Further, ZnONPs have also been used as an effective carrier for the targeted delivery of several anticancer drugs into tumor cells. The increasing focus on ZnONPs resulted in the development of various synthesis approaches including chemical, pHysical, and green or biological for the manufacturing of ZnONPs. In this article, at first we have discussed the various synthesis methods of ZnONPs and secondly its biomedical applications. We have extensively reviewed the anticancer mechanism of ZnONPs on different types of cancers considering its size, shape and surface charge dependent cytotoxicity. Photoirradiation with UV light or NIR laser further increase its anticancer activity via synergistic chemo-photodynamic effect. The drug delivery applications of ZnONPs with special emphasis on drug loading mechanism, stimuli-responsive controlled release and therapeutic effects have also been discussed in this review. Finally, its side effects to vital body organs with mechanism via different exposure routes, the future direction of the ZnONPs research and application are also discussed.
Collapse
Affiliation(s)
- Th Abhishek Singh
- Advance School of Chemical Sciences, Faculty of Basic Sciences, Shoolini University, Solan, Himachal Pradesh 173212, India
| | - Joydeep Das
- Advance School of Chemical Sciences, Faculty of Basic Sciences, Shoolini University, Solan, Himachal Pradesh 173212, India.
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700054, India.
| |
Collapse
|
37
|
Chen Y, Yang J, Fu S, Wu J. Gold Nanoparticles as Radiosensitizers in Cancer Radiotherapy. Int J Nanomedicine 2020; 15:9407-9430. [PMID: 33262595 PMCID: PMC7699443 DOI: 10.2147/ijn.s272902] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/22/2020] [Indexed: 12/19/2022] Open
Abstract
The rapid development of nanotechnology offers a variety of potential therapeutic strategies for cancer treatment. High atomic element nanomaterials are often utilized as radiosensitizers due to their unique photoelectric decay characteristics. Among them, gold nanoparticles (GNPs) are one of the most widely investigated and are considered to be an ideal radiosensitizers for radiotherapy due to their high X-ray absorption and unique physicochemical properties. Over the last few decades, multi-disciplinary studies have focused on the design and optimization of GNPs to achieve greater dosing capability and higher therapeutic effects and highlight potential mechanisms for radiosensitization of GNPs. Although the radiosensitizing potential of GNPs has been widely recognized, its clinical translation still faces many challenges. This review analyses the different roles of GNPs as radiosensitizers in cancer radiotherapy and summarizes recent advances. In addition, the underlying mechanisms of GNP radiosensitization, including physical, chemical and biological mechanisms are discussed, which may provide new directions for the optimization and clinical transformation of next-generation GNPs.
Collapse
Affiliation(s)
- Yao Chen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, People's Republic of China
| | - Juan Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, People's Republic of China
| | - Shaozhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, People's Republic of China
| | - Jingbo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, People's Republic of China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan Province, People's Republic of China
| |
Collapse
|
38
|
Vilas-Boas V, Vinken M. Hepatotoxicity induced by nanomaterials: mechanisms and in vitro models. Arch Toxicol 2020; 95:27-52. [PMID: 33155068 DOI: 10.1007/s00204-020-02940-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/20/2020] [Indexed: 12/14/2022]
Abstract
The unique physicochemical properties of materials at nanoscale have opened a plethora of opportunities for applications in the pharmaceutical and medical field, but also in consumer products from food and cosmetics industries. As a consequence, daily human exposure to nanomaterials through distinct routes is considerable and, therefore, may raise health concerns. Many nanomaterials have been described to accumulate and induce adversity in the liver. Among these, silica and some types of metallic nanoparticles are the most broadly used in consumer products and, therefore, the most studied and reported. The reviewed literature was collected from PubMed.gov during the month of March 2020 using the search words "nanomaterials induced hepatotoxicity", which yielded 181 papers. This present paper reviews the hepatotoxic effects of nanomaterials described in in vitro and in vivo studies, with emphasis on the underlying mechanisms. The induction of oxidative stress and inflammation are the manifestations of toxicity most frequently reported following exposure of cells or animal models to different nanomaterials. Furthermore, the available in vitro models for the evaluation of the hepatotoxic effects of nanomaterials are discussed, highlighting the continuous interest in the development of more advanced and reliable in vitro models for nanotoxicology.
Collapse
Affiliation(s)
- Vânia Vilas-Boas
- Department of In Vitro Toxicology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Mathieu Vinken
- Department of In Vitro Toxicology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
| |
Collapse
|
39
|
Ma R, Qi Y, Zhao X, Li X, Sun X, Niu P, Li Y, Guo C, Chen R, Sun Z. Amorphous silica nanoparticles accelerated atherosclerotic lesion progression in ApoE -/- mice through endoplasmic reticulum stress-mediated CD36 up-regulation in macrophage. Part Fibre Toxicol 2020; 17:50. [PMID: 33008402 PMCID: PMC7531166 DOI: 10.1186/s12989-020-00380-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/14/2020] [Indexed: 01/10/2023] Open
Abstract
Background The biosafety concern of silica nanoparticles (SiNPs) is rapidly expanding alongside with its mass production and extensive applications. The cardiovascular effects of SiNPs exposure have been gradually confirmed, however, the interaction between SiNPs exposure and atherosclerosis, and the underlying mechanisms still remain unknown. Thereby, this study aimed to explore the effects of SiNPs on the progression of atherosclerosis, and to investigate related mechanisms. Results We firstly investigated the in vivo effects of SiNPs exposure on atherosclerosis via intratracheal instillation of ApoE−/− mice fed a Western diet. Ultrasound microscopy showed a significant increase of pulse wave velocity (PWV) compared to the control group, and the histopathological investigation reflected a greater plaque burden in the aortic root of SiNPs-exposed ApoE−/− mice. Compared to the control group, the serum levels of total triglycerides (TG) and low-density lipoprotein cholesterol (LDL-C) were elevated after SiNPs exposure. Moreover, intensified macrophage infiltration and endoplasmic reticulum (ER) stress was occurred in plaques after SiNPs exposure, as evidenced by the upregulated CD68 and CHOP expressions. Further in vitro, SiNPs was confirmed to activate ER stress and induce lipid accumulation in mouse macrophage, RAW264.7. Mechanistic analyses showed that 4-PBA (a classic ER stress inhibitor) pretreatment greatly alleviated SiNPs-induced macrophage lipid accumulation, and reversed the elevated CD36 expression induced by SiNPs. Conclusions Our results firstly revealed the acceleratory effect of SiNPs on the progression of atherosclerosis in ApoE−/− mice, which was related to lipid accumulation caused by ER stress-mediated upregulation of CD36 expression in macrophage. Graphical abstract ![]()
Collapse
Affiliation(s)
- Ru Ma
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Yi Qi
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Xinying Zhao
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.,Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Xueyan Li
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Xuejing Sun
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Piye Niu
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Yanbo Li
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China. .,Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China.
| | - Caixia Guo
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China. .,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| | - Rui Chen
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.,Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Zhiwei Sun
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.,Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
40
|
Anticancer Properties of Platinum Nanoparticles and Retinoic Acid: Combination Therapy for the Treatment of Human Neuroblastoma Cancer. Int J Mol Sci 2020; 21:ijms21186792. [PMID: 32947930 PMCID: PMC7554966 DOI: 10.3390/ijms21186792] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/09/2020] [Accepted: 09/11/2020] [Indexed: 12/13/2022] Open
Abstract
Neuroblastoma is the most common extracranial solid tumor in childhood. The different treatments available for neuroblastoma are challenged by high rates of resistance, recurrence, and progression, most notably in advanced cases and highly malignant tumors. Therefore, the development of more targeted therapies, which are biocompatible and without undesired side effects, is highly desirable. The mechanisms of actions of platinum nanoparticles (PtNPs) and retinoic acid (RA) in neuroblastoma have remained unclear. In this study, the anticancer effects of PtNPs and RA on neuroblastoma were assessed. We demonstrated that treatment of SH-SY5Y cells with the combination of PtNPs and RA resulted in improved anticancer effects. The anticancer effects of the two compounds were mediated by cytotoxicity, oxidative stress (OS), mitochondrial dysfunction, endoplasmic reticulum stress (ERS), and apoptosis-associated networks. Cytotoxicity was confirmed by leakage of lactate dehydrogenase (LDH) and intracellular protease, and oxidative stress increased the level of reactive oxygen species (ROS), 4-hydroxynonenal (HNE), malondialdehyde (MDA), and nitric oxide (NO), and protein carbonyl content (PCC). The combination of PtNPs and RA caused mitochondrial dysfunction by decreasing the mitochondrial membrane potential (MMP), adenosine triphosphate (ATP) content, number of mitochondria, and expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α). Endoplasmic reticulum-mediated stress and apoptosis were confirmed by upregulation of protein kinase RNA-like endoplasmic reticulum kinase (PERK), inositol-requiring enzyme 1 (IRE1), activating transcription factor 6 (ATF6), activating transcription factor 4 (ATF4), p53, Bax, and caspase-3 and down regulation of B-cell lymphoma 2 (BCl-2). PtNPs and RA induced apoptosis, and oxidative DNA damage was evident by the accumulation of 8-hydroxy-2-deoxyguanosine (8-OHdG) and 8-hydroxyguanosine (8-OHG). Finally, PtNPs and RA increased the differentiation and expression of differentiation markers. Differentiated SH-SY5Y cells pre-treated with PtNPs or RA or the combination of both were more sensitive to the cytotoxic effect of cisplatin than undifferentiated cells. To our knowledge, this is the first study to demonstrate the effect of the combination of PtNPs and RA in neuroblastoma cells. PtNPs may be a potential preconditioning or adjuvant compound in chemotherapeutic treatment. The results of this study provide a rationale for clinical evaluation of the combination of PtNPs and RA for the treatment of children suffering from high-risk neuroblastoma.
Collapse
|
41
|
Liao C, Jin Y, Li Y, Tjong SC. Interactions of Zinc Oxide Nanostructures with Mammalian Cells: Cytotoxicity and Photocatalytic Toxicity. Int J Mol Sci 2020; 21:E6305. [PMID: 32878253 PMCID: PMC7504403 DOI: 10.3390/ijms21176305] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022] Open
Abstract
This article presents a state-of-the-art review and analysis of literature studies on the morphological structure, fabrication, cytotoxicity, and photocatalytic toxicity of zinc oxide nanostructures (nZnO) of mammalian cells. nZnO with different morphologies, e.g., quantum dots, nanoparticles, nanorods, and nanotetrapods are toxic to a wide variety of mammalian cell lines due to in vitro cell-material interactions. Several mechanisms responsible for in vitro cytotoxicity have been proposed. These include the penetration of nZnO into the cytoplasm, generating reactive oxygen species (ROS) that degrade mitochondrial function, induce endoplasmic reticulum stress, and damage deoxyribonucleic acid (DNA), lipid, and protein molecules. Otherwise, nZnO dissolve extracellularly into zinc ions and the subsequent diffusion of ions into the cytoplasm can create ROS. Furthermore, internalization of nZnO and localization in acidic lysosomes result in their dissolution into zinc ions, producing ROS too in cytoplasm. These ROS-mediated responses induce caspase-dependent apoptosis via the activation of B-cell lymphoma 2 (Bcl2), Bcl2-associated X protein (Bax), CCAAT/enhancer-binding protein homologous protein (chop), and phosphoprotein p53 gene expressions. In vivo studies on a mouse model reveal the adverse impacts of nZnO on internal organs through different administration routes. The administration of ZnO nanoparticles into mice via intraperitoneal instillation and intravenous injection facilitates their accumulation in target organs, such as the liver, spleen, and lung. ZnO is a semiconductor with a large bandgap showing photocatalytic behavior under ultraviolet (UV) light irradiation. As such, photogenerated electron-hole pairs react with adsorbed oxygen and water molecules to produce ROS. So, the ROS-mediated selective killing for human tumor cells is beneficial for cancer treatment in photodynamic therapy. The photoinduced effects of noble metal doped nZnO for creating ROS under UV and visible light for killing cancer cells are also addressed.
Collapse
Affiliation(s)
- Chengzhu Liao
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; (C.L.); (Y.J.)
| | - Yuming Jin
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; (C.L.); (Y.J.)
| | - Yuchao Li
- Department of Materials Science and Engineering, Liaocheng University, Liaocheng 252000, China
| | - Sie Chin Tjong
- Department of Physics, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| |
Collapse
|
42
|
Qin J, Ru S, Wang W, Hao L, Ru Y, Wang J, Zhang X. Long-term bisphenol S exposure aggravates non-alcoholic fatty liver by regulating lipid metabolism and inducing endoplasmic reticulum stress response with activation of unfolded protein response in male zebrafish. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 263:114535. [PMID: 32283406 DOI: 10.1016/j.envpol.2020.114535] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 03/31/2020] [Accepted: 04/03/2020] [Indexed: 06/11/2023]
Abstract
Environmental chemical exposures have been implicated as risk factors for the development of non-alcoholic fatty liver (NAFLD). Bisphenol S (BPS), widely used in multitudinous consumer products, could disrupt lipid metabolism in the liver. This study aimed at examining the hypothesis that long-term exposure to BPS promotes the development of liver fibrosis and inflammation by means of the application of a semi-static exposure experiment that exposed zebrafish to 1, 10, and 100 μg/L BPS from 3 h post fertilization to 120 day post fertilization. Results showed that the 120-d BPS exposure elevated plasma aspartate aminotransferase and alanine aminotransferase activities, increased triacylglycerol (TAG) and total cholesterol levels in male liver, and even induced hepatic apoptosis and fibrosis. Hepatic lipid accumulation observed in the 30-d BPS-exposed zebrafish was recovered after a 90-d depuration phase, thereby indicating that long-term BPS exposure promotes the progression of simple steatosis to non-alcoholic steatohepatitis. Furthermore, BPS exposure for 120-d promoted the synthesis of TAG and lipotoxic free fatty acids by elevating the transcription of srebp1, acc, fasn, and elovl6, induced endoplasmic reticulum (ER) stress with increasing expression levels of unfolded protein response (UPR) genes (perk, hsp5, atf4a, and ddit3), and then stimulated the expression of two key autophagy genes (atg3 and lc3) and inflammatory genes (il1b and tnfα). It is indicated that BPS can induce the development of steatohepatitis via the activation of the PERK-ATF4a pathway of the UPR. Data gathered suggest that environmental pollutants-induced ER stress with the activation of UPR can potentially trigger the NAFLD development in males. Overall, our study provided new sights into understanding of the adverse health effects of metabolism disrupting chemicals.
Collapse
Affiliation(s)
- Jingyu Qin
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Shaoguo Ru
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Weiwei Wang
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Liping Hao
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Yiran Ru
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, 92093, USA
| | - Jun Wang
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Xiaona Zhang
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
43
|
Quan JH, Gao FF, Lee M, Yuk JM, Cha GH, Chu JQ, Wang H, Lee YH. Involvement of endoplasmic reticulum stress response and IRE1-mediated ASK1/JNK/Mcl-1 pathways in silver nanoparticle-induced apoptosis of human retinal pigment epithelial cells. Toxicology 2020; 442:152540. [PMID: 32717251 DOI: 10.1016/j.tox.2020.152540] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/12/2020] [Accepted: 07/21/2020] [Indexed: 10/23/2022]
Abstract
Silver nanoparticles (AgNPs) have cytotoxic effects on various human cell types. The endoplasmic reticulum (ER) is very sensitive to cytotoxic damage. Retina tissue is easily affected by internal and external stimuli. However, the effect of AgNPs on human retinal cells is not known. This study examined the effect of AgNPs on ER stress induction and their mechanism of action in human retinal pigment epithelium (RPE) ARPE-19 cells. We found that AgNPs significantly increased ARPE-19 cell cytotoxicity and stimulated caspase-3 and poly (ADP-ribose) polymerase (PARP) cleavage, as well as mitochondrial membrane potential (MMP) depolarization, in ARPE-19 cells in a dose-dependent manner (0.2-5 μg/mL for 18 h). AgNPs (5 μg/mL for 18 h) induced several signature ER stress markers, as indicated by the upregulated expressions of CCAAT/enhancer-binding protein-homologous protein (CHOP), phosphorylated protein kinase RNA-like ER kinase (PERK), eukaryotic initiation factor 2α (eIF2α), and inositol-requiring protein 1 (IRE1), and cleaved activating transcription factor 6 (ATF6). AgNPs also activated ASK1 and JNK in ARPE-19 cells, and induced increases in Bax and Puma expressions, as well as a decrease in Mcl-1 expression. However, inhibition of the ER stress response by pretreatment with 4-PBA included apparently and dose-dependently reduced levels of p-PERK, p-IRE1, CHOP, cleaved ATF6, p-ASK1, p-JNK, cleaved caspase-3, procaspase-12, and MMP depolarization in AgNP-treated ARPE-19 cells; it also led to significantly increased Mcl-1 protein levels in a dose-dependent manner in ARPE-19 cells. Pretreatment with JNK inhibitor SP600125 significantly attenuated caspase-3 cleavage and MMP depolarization and increased Mcl-1 protein levels in AgNPs-treated ARPE-19 cells in a dose-dependent manner. Hence, our study demonstrated that AgNPs induced apoptosis in human RPE ARPE-19 cells by ER stress response and ER stress-dependent mitochondrial apoptosis via the IRE1/ASK1/JNK/Mcl-1 pathways.
Collapse
Affiliation(s)
- Juan-Hua Quan
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Fei Fei Gao
- Department of Infection Biology and Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea
| | - Mina Lee
- Department of Obstetrics and Gynecology, Chungnam National University, 33, Munhwa-ro, Jung-gu, Deajeon 35015, Republic of Korea
| | - Jae-Min Yuk
- Department of Infection Biology and Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea
| | - Guang-Ho Cha
- Department of Infection Biology and Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea
| | - Jia-Qi Chu
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Hao Wang
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China.
| | - Young-Ha Lee
- Department of Infection Biology and Department of Medical Science, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea.
| |
Collapse
|
44
|
Malaviya P, Shukal D, Vasavada AR. Nanotechnology-based Drug Delivery, Metabolism and Toxicity. Curr Drug Metab 2020; 20:1167-1190. [PMID: 31902350 DOI: 10.2174/1389200221666200103091753] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/02/2019] [Accepted: 11/23/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Nanoparticles (NPs) are being used extensively owing to their increased surface area, targeted delivery and enhanced retention. NPs have the potential to be used in many disease conditions. Despite widespread use, their toxicity and clinical safety still remain a major concern. OBJECTIVE The purpose of this study was to explore the metabolism and toxicological effects of nanotherapeutics. METHODS Comprehensive, time-bound literature search was done covering the period from 2010 till date. The primary focus was on the metabolism of NP including their adsorption, degradation, clearance, and bio-persistence. This review also focuses on updated investigations on NPs with respect to their toxic effects on various in vitro and in vivo experimental models. RESULTS Nanotechnology is a thriving field of biomedical research and an efficient drug delivery system. Further their applications are under investigation for diagnosis of disease and as medical devices. CONCLUSION The toxicity of NPs is a major concern in the application of NPs as therapeutics. Studies addressing metabolism, side-effects and safety of NPs are desirable to gain maximum benefits of nanotherapeutics.
Collapse
Affiliation(s)
- Pooja Malaviya
- Department of Cell and Molecular Biology, Iladevi Cataract and IOL Research Centre, Memnagar, Ahmedabad 380052, India.,Ph.D. Scholars, Manipal Academy of Higher Education, Manipal, India
| | - Dhaval Shukal
- Department of Cell and Molecular Biology, Iladevi Cataract and IOL Research Centre, Memnagar, Ahmedabad 380052, India.,Ph.D. Scholars, Manipal Academy of Higher Education, Manipal, India
| | - Abhay R Vasavada
- Department of Cell and Molecular Biology, Iladevi Cataract and IOL Research Centre, Memnagar, Ahmedabad 380052, India
| |
Collapse
|
45
|
Rana SVS. Endoplasmic Reticulum Stress Induced by Toxic Elements-a Review of Recent Developments. Biol Trace Elem Res 2020; 196:10-19. [PMID: 31686395 DOI: 10.1007/s12011-019-01903-3] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 09/12/2019] [Indexed: 12/13/2022]
Abstract
Endoplasmic reticulum of all eukaryotic cells is a membrane-bound organelle. Under electron microscope it appears as parallel arrays of "rough membranes" and a maze of "smooth vesicles" respectively. It performs various functions in cell, i.e., synthesis of proteins to degradation of xenobiotics. Bioaccumulation of drugs/chemicals/xenobiotics in the cytosol can trigger ER stress. It is recognized by the accumulation of unfolded or misfolded proteins in the lumen of ER. Present review summarizes the present status of knowledge on ER stress caused by toxic elements, viz arsenic, cadmium, lead, mercury, copper, chromium, and nickel. While inorganic arsenic may induce various glucose-related proteins, i.e., GRP78, GRP94 and CHOP, XBP1, and calpains, cadmium upregulates GRP78. Antioxidants like ascorbic acid, NAC, and Se inhibit the expression of UPR. Exposure to lead also changes ER stress related genes, i.e., GRP 78, GRP 94, ATF4, and ATF6. Mercury too upregulates these genes. Nickel, a carcinogenic element upregulates the expression of Bak, cytochrome C, caspase-3, caspase-9, caspase-12, and GADD 153. Much is not known on ER stress caused by nanoparticles. The review describes inter-organelle association between mitochondria and ER. It also discusses the interdependence between oxidative stress and ER stress. A cross talk amongst different cellular components appears essential to disturb pathways leading to cell death. However, these molecular switches within the signaling network used by toxic elements need to be identified. Nevertheless, ER stress especially caused by toxic elements still remains to be an engaging issue.
Collapse
Affiliation(s)
- S V S Rana
- Department of Toxicology, Ch. Charan Singh University, Meerut, 250 004, India.
| |
Collapse
|
46
|
Ultrafine silicon dioxide nanoparticles cause lung epithelial cells apoptosis via oxidative stress-activated PI3K/Akt-mediated mitochondria- and endoplasmic reticulum stress-dependent signaling pathways. Sci Rep 2020; 10:9928. [PMID: 32555254 PMCID: PMC7303152 DOI: 10.1038/s41598-020-66644-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 04/06/2020] [Indexed: 02/06/2023] Open
Abstract
Silicon dioxide nanoparticles (SiO2NPs) are widely applied in industry, chemical, and cosmetics. SiO2NPs is known to induce pulmonary toxicity. In this study, we investigated the molecular mechanisms of SiO2NPs on pulmonary toxicity using a lung alveolar epithelial cell (L2) model. SiO2NPs, which primary particle size was 12 nm, caused the accumulation of intracellular Si, the decrease in cell viability, and the decrease in mRNAs expression of surfactant, including surfactant protein (SP)-A, SP-B, SP-C, and SP-D. SiO2NPs induced the L2 cell apoptosis. The increases in annexin V fluorescence, caspase-3 activity, and protein expression of cleaved-poly (ADP-ribose) polymerase (PARP), cleaved-caspase-9, and cleaved-caspase-7 were observed. The SiO2NPs induced caspase-3 activity was reversed by pretreatment of caspase-3 inhibitor Z-DEVD-FMK. SiO2NPs exposure increased reactive oxygen species (ROS) production, decreased mitochondrial transmembrane potential, and decreased protein and mRNA expression of Bcl-2 in L2 cells. SiO2NPs increased protein expression of cytosolic cytochrome c and Bax, and mRNAs expression of Bid, Bak, and Bax. SiO2NPs could induce the endoplasmic reticulum (ER) stress-related signals, including the increase in CHOP, XBP-1, and phospho-eIF2α protein expressions, and the decrease in pro-caspase-12 protein expression. SiO2NPs increased phosphoinositide 3-kinase (PI3K) activity and AKT phosphorylation. Both ROS inhibitor N-acetyl-l-cysteine (NAC) and PI3K inhibitor LY294002 reversed SiO2NPs-induced signals described above. However, the LY294002 could not inhibit SiO2NPs-induced ROS generation. These findings demonstrated first time that SiO2NPs induced L2 cell apoptosis through ROS-regulated PI3K/AKT signaling and its downstream mitochondria- and ER stress-dependent signaling pathways.
Collapse
|
47
|
Wu H, Guo H, Liu H, Cui H, Fang J, Zuo Z, Deng J, Li Y, Wang X, Zhao L. Copper sulfate-induced endoplasmic reticulum stress promotes hepatic apoptosis by activating CHOP, JNK and caspase-12 signaling pathways. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 191:110236. [PMID: 32001424 DOI: 10.1016/j.ecoenv.2020.110236] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/16/2020] [Accepted: 01/21/2020] [Indexed: 06/10/2023]
Abstract
Copper (Cu), a transition metal, is an essential trace element in human and animal nutrition at low concentration, but Cu has toxic effects on tissues and organs at high concentration. Endoplasmic reticulum (ER) is a toxicological target in Cu poison. Thus far, no studies have focused on the relationship among copper, endoplasmic reticulum (ER) stress and apoptosis in animal and human livers. In the present study, mice treated with copper sulfate (CuSO4) were used to assess the impacts of copper on ER stress and hepatic apoptosis. A total of 240 mice were orally administered with 0 (control), 10, 20 and 40 mg/kg of CuSO4 for 42 days. The results indicated that CuSO4 at 10 mg/kg markedly induced hepatocyte apoptosis and ER stress. In addition, ER stress was characterized by the increased mRNA and protein levels of glucose-regulated protein 78 (GRP78) and 94 (GRP94). Furthermore, ER stress-triggered 3 apoptotic pathways were also activated by the increased intracellular calcium and up-regulated expression levels of genes involved in growth arrest- and DNA damage-inducible gene 153 (Gadd153/CHOP), c-Jun N-terminal kinase (JNK) and cysteine aspartate-specific protease 12 (caspase-12) signaling pathways in CuSO4-treated mice. In conclusion, CuSO4-induced ER stress can promote hepatic apoptosis in mice by activating CHOP, JNK and caspase-12 signaling pathways.
Collapse
Affiliation(s)
- Hongbin Wu
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Hongrui Guo
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| | - Huan Liu
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Hengmin Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Agricultural information engineering of Sichuan Province, Sichuan Agriculture University, Yaan, Sichuan, 625014, China.
| | - Jing Fang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| | - Zhicai Zuo
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| | - Junliang Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| | - Yinglun Li
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| | - Xun Wang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| | - Ling Zhao
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China; Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, 611130, China
| |
Collapse
|
48
|
Persaud I, Raghavendra AJ, Paruthi A, Alsaleh NB, Minarchick VC, Roede JR, Podila R, Brown JM. Defect-induced electronic states amplify the cellular toxicity of ZnO nanoparticles. Nanotoxicology 2020; 14:145-161. [PMID: 31553248 PMCID: PMC7036006 DOI: 10.1080/17435390.2019.1668067] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 12/31/2022]
Abstract
Zinc oxide nanoparticles (ZnO NPs) are used in numerous applications, including sunscreens, cosmetics, textiles, and electrical devices. Increased consumer and occupational exposure to ZnO NPs potentially poses a risk for toxicity. While many studies have examined the toxicity of ZnO NPs, little is known regarding the toxicological impact of inherent defects arising from batch-to-batch variations. It was hypothesized that the presence of varying chemical defects in ZnO NPs will contribute to cellular toxicity in rat aortic endothelial cells (RAECs). Pristine and defected ZnO NPs (oxidized, reduced, and annealed) were prepared and assessed three major cellular outcomes; cytotoxicity/apoptosis, reactive oxygen species production and oxidative stress, and endoplasmic reticulum (ER) stress. ZnO NPs chemical defects were confirmed by X-ray photoelectron spectroscopy and photoluminescence. Increased toxicity was observed in defected ZnO NPs compared to the pristine NPs as measured by cell viability, ER stress, and glutathione redox potential. It was determined that ZnO NPs induced ER stress through the PERK pathway. Taken together, these results demonstrate a previously unrecognized contribution of chemical defects to the toxicity of ZnO NPs, which should be considered in the risk assessment of engineered nanomaterials.
Collapse
Affiliation(s)
- Indushekhar Persaud
- Colorado Center for Nanomedicine and Nanosafety, Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Achyut J. Raghavendra
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
- Clemson Nanomaterials Center and COMSET, Clemson University, Anderson, SC 29625, USA
| | - Archini Paruthi
- Colorado Center for Nanomedicine and Nanosafety, Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Materials Science and Engineering, Indian Institute of Technology, Gandhinagar, Gujarat, 382355, IN
| | - Nasser B. Alsaleh
- Colorado Center for Nanomedicine and Nanosafety, Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Valerie C. Minarchick
- Colorado Center for Nanomedicine and Nanosafety, Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - James R. Roede
- Colorado Center for Nanomedicine and Nanosafety, Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ramakrishna Podila
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
- Clemson Nanomaterials Center and COMSET, Clemson University, Anderson, SC 29625, USA
| | - Jared M. Brown
- Colorado Center for Nanomedicine and Nanosafety, Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
49
|
Yang Y, Song Z, Wu W, Xu A, Lv S, Ji S. ZnO Quantum Dots Induced Oxidative Stress and Apoptosis in HeLa and HEK-293T Cell Lines. Front Pharmacol 2020; 11:131. [PMID: 32180717 PMCID: PMC7057716 DOI: 10.3389/fphar.2020.00131] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 01/30/2020] [Indexed: 12/17/2022] Open
Abstract
Zinc oxide (ZnO) quantum dot (QD) is a promising inexpensive inorganic nanomaterials, of which potential toxic effects on biological systems and human health should be evaluated before biomedical application. In this study, the cytotoxicity of ZnO QDs was assessed using HeLa cervical cancer cell and HEK-293T human embryonic kidney cell lines. Cell viability was significantly decreased by treatment with 50 µg/ml ZnO QDs after only 6 h, and the cytotoxicity of ZnO QDs was higher in HEK-293T than in HeLa cells. ZnO QDs increased the level of reactive oxygen species and decreased the mitochondria membrane potential in a dose-dependent manner. Several gene expression involved in apoptosis was regulated by ZnO QDs, including bcl-2 gene and caspase. In HeLa cells, ZnO QDs significantly increased early and late apoptosis, but only late apoptosis was affected in HEK-293T cells. These findings will be helpful for future research and application of ZnO QDs in biomedicine.
Collapse
Affiliation(s)
- Yanjie Yang
- Provincial Engineering Centre for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Zhenhua Song
- Provincial Engineering Centre for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Weixia Wu
- Provincial Engineering Centre for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Ao Xu
- Provincial Engineering Centre for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Shuangyu Lv
- Provincial Engineering Centre for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Shaoping Ji
- Provincial Engineering Centre for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| |
Collapse
|
50
|
|