1
|
Liu N, Zhang B, Lin N. Review on the role of autophagy in the toxicity of nanoparticles and the signaling pathways involved. Chem Biol Interact 2025; 406:111356. [PMID: 39701490 DOI: 10.1016/j.cbi.2024.111356] [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: 10/27/2024] [Revised: 12/09/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
As the development of nanotechnology, the application of nanoproducts and the advancement of nanomedicine, the contact of nanoparticles (NPs) with human body is becoming increasingly prevalent. This escalation elevates the risk of NPs exposure for workers, consumers, researchers, and both aquatic and terrestrial organisms throughout the production, usage, and disposal stages. Consequently, evaluating nanotoxicity remains critically important, though standardized assessment criteria are still lacking. The diverse and complex properties of NPs further complicate the understanding of their toxicological mechanisms. Autophagy, a fundamental cellular process, exhibits dual functions-both pro-survival and pro-death. This review offers an updated perspective on the dual roles of autophagy in nanotoxicity and examines the factors influencing autophagic responses. However, no definitive framework exists for predicting NPs-induced autophagy. Beyond the conventional autophagy pathways, the review highlights specific transcription factors activated by NPs and explores metabolic reprogramming. Particular attention is given to NPs-induced selective autophagy, including mitophagy, ER-phagy, ferritinophagy, lysophagy, and lipophagy. Additionally, the review investigates autophagy's involvement in NPs-mediated biological processes such as ferroptosis, inflammation, macrophage polarization, epithelial-mesenchymal transition, tumor cell proliferation and drug resistance, as well as liver and kidney injury, neurotoxicity, and other diseases. In summary, this review presents a novel update on selective autophagy-mediated nanotoxicity and elucidates the broader interactions of autophagy in NPs-induced biological processes. Collectively, these insights offer valuable strategies for mitigating nanotoxicity through autophagy modulation and advancing the development of NPs in biomedical applications.
Collapse
Affiliation(s)
- Na Liu
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Bo Zhang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China.
| | - Nengming Lin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China.
| |
Collapse
|
2
|
Li J, Yuan Z, Shi S, Chen X, Yu S, Qi X, Deng T, Zhou Y, Tang D, Xu S, Zhang J, Jiao Y, Yu W, Wang L, Yang L, Gao P. Microneedle patches incorporating zinc-doped mesoporous silica nanoparticles loaded with betamethasone dipropionate for psoriasis treatment. J Nanobiotechnology 2024; 22:706. [PMID: 39543615 PMCID: PMC11562306 DOI: 10.1186/s12951-024-02986-4] [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: 08/01/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
Treating psoriasis presents a major clinical challenge because of the limitations associated with traditional topical glucocorticoid therapy. This study introduced a drug delivery system utilizing zinc-doped mesoporous silica nanoparticle (Zn-MSN) and microneedle (MN), designed to enhance drug utilization for prolonged anti-inflammatory and anti-itch effects. The MN system facilitated the transdermal delivery of betamethasone dipropionate (BD), allowing its slow release. The BD@Zn-MSN-MN system promoted the polarization of macrophages towards the anti-inflammatory M2 phenotype, achieving superior anti-inflammatory effects compared to the clinically used BD cream. Additionally, this study demonstrated that BD@Zn-MSN-MN could further alleviate itching in psoriasis-afflicted mice by decreasing the excitability of the transient receptor potential vanilloid V1 (TRPV1) ion channel positive neurons and reducing the release of calcitonin gene-related peptide (CGRP) in the dorsal root ganglion (DRG). These findings offer new insights and effective therapeutic options for the future design of transdermal drug delivery for psoriasis.
Collapse
Grants
- 20DZ2254200 Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation
- 20DZ2254200 Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation
- 20DZ2254200 Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation
- 20DZ2254200 Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation
- 20DZ2254200 Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation
- 20DZ2254200 Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation
- 20DZ2254200 Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation
- 20DZ2254200 Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation
- 20DZ2254200 Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation
- 20DZ2254200 Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation
- 20DZ2254200 Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation
- 20DZ2254200 Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation
- 20DZ2254200 Shanghai Engineering Research Center of Peri-operative Organ Support and Function Preservation
- 2023AH010073 Program for Excellent Sci-tech Innovation Teams of Universities in Anhui Province
- 2023AH010073 Program for Excellent Sci-tech Innovation Teams of Universities in Anhui Province
- 82270916, U23A20508, 82371517, 32030043, 81800748 National Natural Science Foundation of China
- 82270916, U23A20508, 82371517, 32030043, 81800748 National Natural Science Foundation of China
- 82270916, U23A20508, 82371517, 32030043, 81800748 National Natural Science Foundation of China
- 82270916, U23A20508, 82371517, 32030043, 81800748 National Natural Science Foundation of China
- 82270916, U23A20508, 82371517, 32030043, 81800748 National Natural Science Foundation of China
- PW2022D-01 Pudong New Area Health Commission Research Project
Collapse
Affiliation(s)
- Jun Li
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Zhiguo Yuan
- Department of Bone and Joint Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Shuyu Shi
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Xingtao Chen
- Sichuan Provincial Laboratory of Orthopedic Engineering, Department of Orthopedics, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Shuangshuang Yu
- Department of Dermatology, Chaohu Hospital Affiliated to Anhui Medical University, Chaohu, Anhui, 238000, China
| | - Xiaoshu Qi
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Tong Deng
- Anhui Province Engineering Research Center for Dental Materials and Application, School of Stomatology, Wannan Medical College, Wuhu, 241002, China
| | - Yifei Zhou
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Dan Tang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Saihong Xu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Jue Zhang
- Anhui Province Engineering Research Center for Dental Materials and Application, School of Stomatology, Wannan Medical College, Wuhu, 241002, China
| | - Yingfu Jiao
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Weifeng Yu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China.
| | - Liya Wang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China.
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, 200030, China.
| | - Liqun Yang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China.
| | - Po Gao
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China.
| |
Collapse
|
3
|
Zhou X, Medina-Ramirez IE, Su G, Liu Y, Yan B. All Roads Lead to Rome: Comparing Nanoparticle- and Small Molecule-Driven Cell Autophagy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310966. [PMID: 38616767 DOI: 10.1002/smll.202310966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/27/2024] [Indexed: 04/16/2024]
Abstract
Autophagy, vital for removing cellular waste, is triggered differently by small molecules and nanoparticles. Small molecules, like rapamycin, non-selectively activate autophagy by inhibiting the mTOR pathway, which is essential for cell regulation. This can clear damaged components but may cause cytotoxicity with prolonged use. Nanoparticles, however, induce autophagy, often causing oxidative stress, through broader cellular interactions and can lead to a targeted form known as "xenophagy." Their impact varies with their properties but can be harnessed therapeutically. In this review, the autophagy induced by nanoparticles is explored and small molecules across four dimensions: the mechanisms behind autophagy induction, the outcomes of such induction, the toxicological effects on cellular autophagy, and the therapeutic potential of employing autophagy triggered by nanoparticles or small molecules. Although small molecules and nanoparticles each induce autophagy through different pathways and lead to diverse effects, both represent invaluable tools in cell biology, nanomedicine, and drug discovery, offering unique insights and therapeutic opportunities.
Collapse
Affiliation(s)
- Xiaofei Zhou
- College of Science & Technology, Hebei Agricultural University, Baoding, 071001, China
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Baoding, 071100, China
| | - Iliana E Medina-Ramirez
- Department of Chemistry, Universidad Autónoma de Aguascalientes, Av Universidad 940, Aguascalientes, Aguascalientes, México
| | - Gaoxing Su
- School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Yin Liu
- School of Environment, Hangzhou Institute for Advanced Study, UCAS, Hangzhou, 10024, China
| | - Bing Yan
- Institute of Environmental Research at the Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou, 510006, China
| |
Collapse
|
4
|
Tang B, Xie X, Lu J, Huang W, Yang J, Tian J, Lei L. Designing biomaterials for the treatment of autoimmune diseases. APPLIED MATERIALS TODAY 2024; 39:102278. [DOI: 10.1016/j.apmt.2024.102278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
|
5
|
Florance I, Cordani M, Pashootan P, Moosavi MA, Zarrabi A, Chandrasekaran N. The impact of nanomaterials on autophagy across health and disease conditions. Cell Mol Life Sci 2024; 81:184. [PMID: 38630152 PMCID: PMC11024050 DOI: 10.1007/s00018-024-05199-y] [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: 11/02/2023] [Revised: 03/01/2024] [Accepted: 03/03/2024] [Indexed: 04/19/2024]
Abstract
Autophagy, a catabolic process integral to cellular homeostasis, is constitutively active under physiological and stress conditions. The role of autophagy as a cellular defense response becomes particularly evident upon exposure to nanomaterials (NMs), especially environmental nanoparticles (NPs) and nanoplastics (nPs). This has positioned autophagy modulation at the forefront of nanotechnology-based therapeutic interventions. While NMs can exploit autophagy to enhance therapeutic outcomes, they can also trigger it as a pro-survival response against NP-induced toxicity. Conversely, a heightened autophagy response may also lead to regulated cell death (RCD), in particular autophagic cell death, upon NP exposure. Thus, the relationship between NMs and autophagy exhibits a dual nature with therapeutic and environmental interventions. Recognizing and decoding these intricate patterns are essential for pioneering next-generation autophagy-regulating NMs. This review delves into the present-day therapeutic potential of autophagy-modulating NMs, shedding light on their status in clinical trials, intervention of autophagy in the therapeutic applications of NMs, discusses the potency of autophagy for application as early indicator of NM toxicity.
Collapse
Affiliation(s)
- Ida Florance
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040, Madrid, Spain.
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040, Madrid, Spain.
| | - Parya Pashootan
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, P.O Box 14965/161, Tehran, Iran
| | - Mohammad Amin Moosavi
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, P.O Box 14965/161, Tehran, Iran
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, 34396, Turkey
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600 077, India
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan, Taiwan
| | - Natarajan Chandrasekaran
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
6
|
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: 10] [Impact Index Per Article: 5.0] [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
|
7
|
Qiao D, Zhang T, Tang M. Autophagy regulation by inorganic, organic, and organic/inorganic hybrid nanoparticles: Organelle damage, regulation factors, and potential pathways. J Biochem Mol Toxicol 2023; 37:e23429. [PMID: 37409715 DOI: 10.1002/jbt.23429] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 03/30/2023] [Accepted: 06/12/2023] [Indexed: 07/07/2023]
Abstract
The rapid development of nanotechnology requires a more thorough understanding of the potential health effects caused by nanoparticles (NPs). As a programmed cell death, autophagy is one of the biological effects induced by NPs, which maintain intracellular homeostasis by degrading damaged organelles and removing aggregates of defective proteins through lysosomes. Currently, autophagy has been shown to be associated with the development of several diseases. A significant number of research have demonstrated that most NPs can regulate autophagy, and their regulation of autophagy is divided into induction and blockade. Studying the autophagy regulation by NPs will facilitate a more comprehensive understanding of the toxicity of NPs. In this review, we will illustrate the effects of different types of NPs on autophagy, including inorganic NPs, organic NPs, and organic/inorganic hybrid NPs. The potential mechanisms by which NPs regulate autophagy are highlighted, including organelle damage, oxidative stress, inducible factors, and multiple signaling pathways. In addition, we list the factors influencing NPs-regulated autophagy. This review may provide basic information for the safety assessment of NPs.
Collapse
Affiliation(s)
- Dong Qiao
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Ting Zhang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Meng Tang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
8
|
Hosseinpour S, Dai H, Walsh LJ, Xu C. Mesoporous Core-Cone Silica Nanoparticles Can Deliver miRNA-26a to Macrophages to Exert Immunomodulatory Effects on Osteogenesis In Vitro. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:1755. [PMID: 37299658 PMCID: PMC10254425 DOI: 10.3390/nano13111755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023]
Abstract
Nanoparticles can play valuable roles in delivering nucleic acids, including microRNAs (miRNA), which are small, non-coding RNA segments. In this way, nanoparticles may exert post-transcriptional regulatory influences on various inflammatory conditions and bone disorders. This study used biocompatible, core-cone-structured, mesoporous silica nanoparticles (MSN-CC) to deliver miRNA-26a to macrophages in order to influence osteogenesis in vitro. The loaded nanoparticles (MSN-CC-miRNA-26) showed low-level toxicity towards macrophages (RAW 264.7 cells) and were internalized efficiently, causing the reduced expression of pro-inflammatory cytokines, as seen via real-time PCR and cytokine immunoassays. The conditioned macrophages created a favorable osteoimmune environment for MC3T3-E1 preosteoblasts, driving osteogenic differentiation with enhanced osteogenic marker expression, alkaline phosphatase (ALP) production, extracellular matrix formation, and calcium deposition. An indirect co-culture system revealed that direct osteogenic induction and immunomodulation by MSN-CC-miRNA-26a synergistically increased bone production due to the crosstalk between MSN-CC-miRNA-26a-conditioned macrophages and MSN-CC-miRNA-26a-treated preosteoblasts. These findings demonstrate the value of nanoparticle delivery of miR-NA-26a using MSN-CC for suppressing the production of pro-inflammatory cytokines with macrophages and for driving osteogenic differentiation in preosteoblasts via osteoimmune modulation.
Collapse
Affiliation(s)
| | | | | | - Chun Xu
- School of Dentistry, The University of Queensland, Herston, QLD 4006, Australia
| |
Collapse
|
9
|
Li B, Zhang T, Tang M. Toxicity mechanism of nanomaterials: Focus on endoplasmic reticulum stress. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 834:155417. [PMID: 35472346 DOI: 10.1016/j.scitotenv.2022.155417] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/06/2022] [Accepted: 04/17/2022] [Indexed: 06/14/2023]
Abstract
Over the years, although the broad application of nanomaterials has not brought convenience to people's life, growing concern surrounds their safety. Recently, much emphasis has been placed on exploring the toxicity mechanism of nanoparticles. Currently established toxic mechanisms include oxidative stress, inflammatory response, autophagy, and DNA damage. In recent years, endoplasmic reticulum stress (ERS) has gained widespread attention as another toxic mechanism of nanomaterials. It is widely acknowledged that the endoplasmic reticulum (ER) is an important site for protein synthesis, and lipids and Ca+ storage, playing an esseential role in the normal operation of the body functions. When the body's internal environment is damaged, the structure and function of the endoplasmic reticulum are destroyed, leading to a series of biological reactions called endoplasmic reticulum stress (ERS.) This paper reviews the mechanism of ERS in nanomaterial-associated toxicity. The process of ERS and its related unfolded protein response were briefly introduced, summarizing the factors affecting the nanoparticle ability to induce ERS and expounding on the changes of ER morphology after exposure to nanoparticles. Finally, the specific role and molecular mechanism of ERS under the action of different nanoparticles were comprehensively analyzed, including the relationship between ERS and inflammation, oxidative stress, lipid metabolism and apoptosis. This review provides a foothold for future studies on the toxic mechanism of nanoparticles, and provides novel insights into the safe application of nanoparticles and the treatment of diseases.
Collapse
Affiliation(s)
- Binjing Li
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
| | - Ting Zhang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Meng Tang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
10
|
BECLIN-1-Mediated Autophagy Suppresses Silica Nanoparticle-Induced Testicular Toxicity via the Inhibition of Caspase 8-Mediated Cell Apoptosis in Leydig Cells. Cells 2022; 11:cells11121863. [PMID: 35740992 PMCID: PMC9221084 DOI: 10.3390/cells11121863] [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: 04/06/2022] [Revised: 05/28/2022] [Accepted: 06/04/2022] [Indexed: 11/17/2022] Open
Abstract
Accumulation of silica nanoparticles (SNPs) in the testes leads to male reproductive toxicity. However, little is known about the effect and mechanistic insights of SNP-induced autophagy on apoptosis in Leydig cells. In this study, we aimed to verify the role of SNP-induced autophagy in apoptosis and explore the possible underlying mechanism in mouse primary Leydig cells (PLCs). H&E staining showed that SNPs changed the histological structures of the testes, including a reduction in the Leydig cell populations in vivo. CCK-8 assay showed that SNPs decreased cell viability, and flow cytometry showed that SNPs increased cell apoptosis, both in a dose-dependent manner in vitro. Additionally, Western blotting further found that SNPs activated autophagy by an increase in BECLIN-1, ATG16L, and LC3-II levels and promoted the intrinsic pathway of apoptosis by an increase in the BAX/BCL-2 ratio, cleaved the caspase 8 and caspase 3 levels. Furthermore, autophagy decreased SNP-induced apoptosis via regulation of the caspase 8 level combined with rapamycin, 3-methyladenine, and chloroquine. BECLIN-1 depletion increased the caspase 8 level, leading to an increase in SNP-induced cell apoptosis. Collectively, this evidence demonstrates that SNPs activated BECLIN-1-mediated autophagy, which prevented SNP-induced testicular toxicity via the inhibition of caspase 8-mediated cell apoptosis in Leydig cells.
Collapse
|
11
|
Wang T, Zhang Z, Xie M, Li S, Zhang J, Zhou J. Apigenin Attenuates Mesoporous Silica Nanoparticles-Induced Nephrotoxicity by Activating FOXO3a. Biol Trace Elem Res 2022; 200:2793-2806. [PMID: 34448149 DOI: 10.1007/s12011-021-02871-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/04/2021] [Indexed: 11/30/2022]
Abstract
Mesoporous silica nanoparticles (MSNs) are widely used in many biomedical applications and clinical fields. However, the applications of MSNs are limited by their severe toxicity. Apigenin (AG) has demonstrated pharmacological effects with low toxicity. The aim of this study was to clarify the role of AG in the progression of MSNs-induced renal injury. BALB/c mice and NRK-52E cells were exposed to MSNs with or without AG. AG protected mice and NRK-52E cells from the MSNs-induced pathological variations in renal tissues and decreased cell viability. AG significantly reduced the levels of serum blood urea nitrogen (BUN) and serum creatinine (Scr), upregulated the levels of superoxide dismutase (SOD), glutathione (GSH) and catalase (CAT), and improved the pathological changes of the kidney in MSNs-treated mice. The protective effects of AG were associated with its ability to increase the levels of antioxidants, reduce the accumulation of ROS, and inhibit the expression of the inflammatory mediators (TNF-α, IL-6). In addition, AG treatment upregulated the activity of FOXO3a, increased the level of IkBα, and reduced the nuclear translocation of NF-κB, which ultimately alleviated MSNs-induced inflammation. Nuclear FOXO3a translocation also triggered antioxidant gene transcription and protected nephrocyte from oxidative damage. However, knockdown of FOXO3a significantly blocked the protective effects of AG. These findings suggested that AG could be a promising therapeutic strategy for MSNs-induced nephrotoxicity, and this protective effect might be related to the suppression of oxidative stress and inflammation via the FOXO3a/NF-κB pathway.
Collapse
Affiliation(s)
- Tianyang Wang
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, People's Republic of China
| | - Ziwen Zhang
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, People's Republic of China
| | - Minjuan Xie
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, People's Republic of China
| | - Saifeng Li
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, People's Republic of China
| | - Jian Zhang
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, People's Republic of China
| | - Jie Zhou
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, People's Republic of China.
| |
Collapse
|
12
|
Zhang J, Ye R, Grunberger JW, Jin J, Zhang Q, Mohammadpour R, Khurana N, Xu X, Ghandehari H, Chen F. Activation of Autophagy by Low-Dose Silica Nanoparticles Enhances Testosterone Secretion in Leydig Cells. Int J Mol Sci 2022; 23:ijms23063104. [PMID: 35328525 PMCID: PMC8949068 DOI: 10.3390/ijms23063104] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 02/05/2023] Open
Abstract
Silica nanoparticles (SNPs) can cause abnormal spermatogenesis in male reproductive toxicity. However, the toxicity and toxicological mechanisms of SNPs in testosterone synthesis and secretion in Leydig cells are not well known. Therefore, this study aimed to determine the effect and molecular mechanism of low doses of SNPs in testosterone production in Leydig cells. For this, mouse primary Leydig cells (PLCs) were exposed to 100 nm Stöber nonporous spherical SNPs. We observed significant accumulation of SNPs in the cytoplasm of PLCs via transmission electron microscopy (TEM). CCK-8 and flow cytometry assays confirmed that low doses (50 and 100 μg/mL) of SNPs had no significant effect on cell viability and apoptosis, whereas high doses (more than 200 μg/mL) decreased cell viability and increased cell apoptosis in PLCs. Monodansylcadaverine (MDC) staining showed that SNPs caused the significant accumulation of autophagosomes in the cytoplasm of PLCs. SNPs activated autophagy by upregulating microtubule-associated protein light chain 3 (LC3-II) and BCL-2-interacting protein (BECLIN-1) levels, in addition to downregulating sequestosome 1 (SQSTM1/P62) level at low doses. In addition, low doses of SNPs enhanced testosterone secretion and increased steroidogenic acute regulatory protein (StAR) expression. SNPs combined with rapamycin (RAP), an autophagy activator, enhanced testosterone production and increased StAR expression, whereas SNPs combined with 3-methyladenine (3-MA) and chloroquine (CQ), autophagy inhibitors, had an opposite effect. Furthermore, BECLIN-1 depletion inhibited testosterone production and StAR expression. Altogether, our results demonstrate that low doses of SNPs enhanced testosterone secretion via the activation of autophagy in PLCs.
Collapse
Affiliation(s)
- Jinlong Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (J.Z.); (R.Y.); (J.J.); (Q.Z.); (X.X.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Rongrong Ye
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (J.Z.); (R.Y.); (J.J.); (Q.Z.); (X.X.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Jason William Grunberger
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA; (J.W.G.); (R.M.); (N.K.); (H.G.)
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA
| | - Jiaqi Jin
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (J.Z.); (R.Y.); (J.J.); (Q.Z.); (X.X.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Qianru Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (J.Z.); (R.Y.); (J.J.); (Q.Z.); (X.X.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Raziye Mohammadpour
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA; (J.W.G.); (R.M.); (N.K.); (H.G.)
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA
| | - Nitish Khurana
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA; (J.W.G.); (R.M.); (N.K.); (H.G.)
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA
| | - Xianyu Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (J.Z.); (R.Y.); (J.J.); (Q.Z.); (X.X.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Hamidreza Ghandehari
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA; (J.W.G.); (R.M.); (N.K.); (H.G.)
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT 84112, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Fenglei Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; (J.Z.); (R.Y.); (J.J.); (Q.Z.); (X.X.)
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Correspondence: ; Tel.: +86-514-87979030; Fax: +86-514-87972218
| |
Collapse
|
13
|
Stephen S, Gorain B, Choudhury H, Chatterjee B. Exploring the role of mesoporous silica nanoparticle in the development of novel drug delivery systems. Drug Deliv Transl Res 2022; 12:105-123. [PMID: 33604837 DOI: 10.1007/s13346-021-00935-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2021] [Indexed: 10/22/2022]
Abstract
The biocompatible nature of mesoporous silica nanoparticles (MSN) attracted researchers' attention to deliver therapeutic agents in the treatment of various diseases, where their porous nature, high drug loading efficiency, and suitability to functionalize with a specific ligand of MSN helped to obtain the desired outcome. The application of MSN has been extended to deliver small chemicals to large-sized peptides or proteins to fight against complex diseases. Recently, formulation researches with MSN have been progressed for various non-conventional drug delivery systems, including liposome, microsphere, oro-dispersible film, 3D-printed formulation, and microneedle. Low bulk density, retaining mesoporous structure during downstream processing, and lack of sufficient in vivo studies are some of the important issues towards the success of mesoporous silica-based advanced drug delivery systems. The present review has aimed to evaluate the application of MSN in advanced drug delivery systems to critically analyze the role of MSN in the respective formulation over other functionalized polymers. Finally, an outlook on the future direction of MSN-based advanced drug delivery systems has been drawn against the existing challenges with this platform.
Collapse
Affiliation(s)
- Senitta Stephen
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V.L Mehta Road, Vile Parle(W), Mumbai, India
| | - Bapi Gorain
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, 47500, Subang Jaya, Selangor, Malaysia
- Centre for Drug Delivery and Molecular Pharmacology, Faculty of Health and Medical Sciences, Taylor's University, 47500, Subang Jaya, Selangor, Malaysia
| | - Hira Choudhury
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Bukit Jalil , 57000, Kuala Lumpur, Malaysia
- Centre for Bioactive Molecules and Drug Delivery, Institute for Research, Development and Innovation, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Bappaditya Chatterjee
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V.L Mehta Road, Vile Parle(W), Mumbai, India.
| |
Collapse
|
14
|
Mohammapdour R, Ghandehari H. Mechanisms of immune response to inorganic nanoparticles and their degradation products. Adv Drug Deliv Rev 2022; 180:114022. [PMID: 34740764 PMCID: PMC8898339 DOI: 10.1016/j.addr.2021.114022] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 09/24/2021] [Accepted: 10/20/2021] [Indexed: 01/03/2023]
Abstract
Careful assessment of the biological fate and immune response of inorganic nanoparticles is crucial for use of such carriers in drug delivery and other biomedical applications. Many studies have elucidated the cellular and molecular mechanisms of the interaction of inorganic nanoparticles with the components of the immune system. The biodegradation and dissolution of inorganic nanoparticles can influence their ensuing immune response. While the immunological properties of inorganic nanoparticles as a function of their physicochemical properties have been investigated in detail, little attention has been paid to the immune adverse effects towards the degradation products of these nanoparticles. To fill this gap, we herein summarize the cellular mechanisms of immune response to inorganic nanoparticles and their degradation products with specific focus on immune cells. We also accentuate the importance of designing new methods and instruments for the in situ characterization of inorganic nanoparticles in order to assess their safety as a result of degradation. This review further sheds light on factors that need to be considered in the design of safe and effective inorganic nanoparticles for use in delivery of bioactive and imaging agents.
Collapse
Affiliation(s)
- Raziye Mohammapdour
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT, USA; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA.
| | - Hamidreza Ghandehari
- Utah Center for Nanomedicine, University of Utah, Salt Lake City, UT, USA; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
15
|
Hosseinpour S, Walsh LJ, Xu C. Modulating Osteoimmune Responses by Mesoporous Silica Nanoparticles. ACS Biomater Sci Eng 2021; 8:4110-4122. [PMID: 34775744 DOI: 10.1021/acsbiomaterials.1c00899] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The immune response plays an important role in biomaterial-mediated osteogenesis. Nanomaterials may influence immune responses and thereby alter bone regeneration. Mesoporous silica nanoparticles (MSNs) have received much attention for drug delivery and bone regeneration. Recently, immunomodulatory effects of MSNs on osteogenesis have been reported. In this Review, we summarize the osteoimmunomodulation of MSNs, including the effects of MSN characteristics on immune cells and osteogenesis. Impacts of MSNs on immune cells vary according to nanoparticle properties, including surface topography and charge, particle size, and ion release. MSNs with suitable doses can inhibit inflammation and create an immune microenvironment beneficial for bone regeneration by activating immune cells and stimulating cytokine release. Further work is needed to explore and clarify the underlying mechanisms, including crosstalk between various types of immune cells and how to design MSNs to create a suitable immune environment for osteogenesis.
Collapse
Affiliation(s)
- Sepanta Hosseinpour
- School of Dentistry, The University of Queensland, Herston, Queensland 4006, Australia
| | - Laurence J Walsh
- School of Dentistry, The University of Queensland, Herston, Queensland 4006, Australia
| | - Chun Xu
- School of Dentistry, The University of Queensland, Herston, Queensland 4006, Australia
| |
Collapse
|
16
|
Sun L, Sogo Y, Wang X, Ito A. Biosafety of mesoporous silica nanoparticles: a combined experimental and literature study. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:102. [PMID: 34406531 PMCID: PMC8373747 DOI: 10.1007/s10856-021-06582-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/28/2021] [Indexed: 05/03/2023]
Abstract
Mesoporous silica (MS) particles have been explored for various healthcare applications, but universal data about their safety and/or toxicity are yet to be well-established for clinical purposes. Information about general toxicity of hollow MS (HMS) particles and about immunotoxicity of MS particles are significantly lacked. Therefore, acute toxicity and immunotoxicity of HMS particles were experimentally evaluated. A systematic and objective literature study was parallelly performed to analyze the published in vivo toxicity of MS particles. Lethal acute toxicity of MS particles is likely to arise from their physical action after intravenous and intraperitoneal administrations, and only rarely observed after subcutaneous administration. No clear relationship was identified between physicochemical properties of MS particles and lethality as well as maximum tolerated dose with some exceptions. At sub-lethal doses, MS particles tend to accumulate mainly in lung, liver, and spleen. The HMS particles showed lower inflammation-inducing ability than polyinosinic-polycytidylic acid and almost the same allergy-inducing ability as Alum. Finally, the universal lowest observed adverse effect levels were determined as 0.45, 0.81, and 4.1 mg/kg (human equivalent dose) for intravenous, intraperitoneal, and subcutaneous administration of MS particles, respectively. These results could be helpful for determining an appropriate MS particle dose in clinical study.
Collapse
Affiliation(s)
- Lue Sun
- Health and Medical Research Institute, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Yu Sogo
- Health and Medical Research Institute, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan.
| | - Xiupeng Wang
- Health and Medical Research Institute, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| | - Atsuo Ito
- Health and Medical Research Institute, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| |
Collapse
|
17
|
High Surface Area Mesoporous Silica Nanoparticles with Tunable Size in the Sub-Micrometer Regime: Insights on the Size and Porosity Control Mechanisms. Molecules 2021; 26:molecules26144247. [PMID: 34299522 PMCID: PMC8304748 DOI: 10.3390/molecules26144247] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/06/2021] [Accepted: 07/09/2021] [Indexed: 11/17/2022] Open
Abstract
Mesoporous silica nanostructures (MSNs) attract high interest due to their unique and tunable physical chemical features, including high specific surface area and large pore volume, that hold a great potential in a variety of fields, i.e., adsorption, catalysis, and biomedicine. An essential feature for biomedical application of MSNs is limiting MSN size in the sub-micrometer regime to control uptake and cell viability. However, careful size tuning in such a regime remains still challenging. We aim to tackling this issue by developing two synthetic procedures for MSN size modulation, performed in homogenous aqueous/ethanol solution or two-phase aqueous/ethyl acetate system. Both approaches make use of tetraethyl orthosilicate as precursor, in the presence of cetyltrimethylammonium bromide, as structure-directing agent, and NaOH, as base-catalyst. NaOH catalyzed syntheses usually require high temperature (>80 °C) and large reaction medium volume to trigger MSN formation and limit aggregation. Here, a successful modulation of MSNs size from 40 up to 150 nm is demonstrated to be achieved by purposely balancing synthesis conditions, being able, in addition, to keep reaction temperature not higher than 50 °C (30 °C and 50 °C, respectively) and reaction mixture volume low. Through a comprehensive and in-depth systematic morphological and structural investigation, the mechanism and kinetics that sustain the control of MSNs size in such low dimensional regime are defined, highlighting that modulation of size and pores of the structures are mainly mediated by base concentration, reaction time and temperature and ageing, for the homogenous phase approach, and by temperature for the two-phase synthesis. Finally, an in vitro study is performed on bEnd.3 cells to investigate on the cytotoxicity of the MNSs.
Collapse
|
18
|
Hosseinpour S, Walsh LJ, Xu C. Biomedical application of mesoporous silica nanoparticles as delivery systems: a biological safety perspective. J Mater Chem B 2021; 8:9863-9876. [PMID: 33047764 DOI: 10.1039/d0tb01868f] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The application of mesoporous silica nanoparticles (MSNs) as drug delivery systems to deliver drugs, proteins, and genes has expanded considerably in recent years, using in vitro and animal studies. For future translation to clinical applications, the biological safety aspects of MSNs must be considered carefully. This paper reviews the biosafety of MSNs, examining key issues such as biocompatibility, effects on immune cells and erythrocytes, biodistribution, biodegradation and clearance, and how these vary depending on the effects of the physical and chemical properties of MSNs such as particle size, porosity, morphology, surface charge, and chemical modifications. The future use of MSNs as a delivery system must extend beyond what has been learnt thus far using rodent animal models to encompass larger animals.
Collapse
Affiliation(s)
- Sepanta Hosseinpour
- School of Dentistry, The University of Queensland, Herston, QLD 4006, Australia.
| | - Laurence J Walsh
- School of Dentistry, The University of Queensland, Herston, QLD 4006, Australia.
| | - Chun Xu
- School of Dentistry, The University of Queensland, Herston, QLD 4006, Australia.
| |
Collapse
|
19
|
Li B, Tang M. Research progress of nanoparticle toxicity signaling pathway. Life Sci 2020; 263:118542. [DOI: 10.1016/j.lfs.2020.118542] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/26/2020] [Accepted: 09/28/2020] [Indexed: 01/19/2023]
|
20
|
Feng X, Zhang Y, Zhang C, Lai X, Zhang Y, Wu J, Hu C, Shao L. Nanomaterial-mediated autophagy: coexisting hazard and health benefits in biomedicine. Part Fibre Toxicol 2020; 17:53. [PMID: 33066795 PMCID: PMC7565835 DOI: 10.1186/s12989-020-00372-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Widespread biomedical applications of nanomaterials (NMs) bring about increased human exposure risk due to their unique physicochemical properties. Autophagy, which is of great importance for regulating the physiological or pathological activities of the body, has been reported to play a key role in NM-driven biological effects both in vivo and in vitro. The coexisting hazard and health benefits of NM-mediated autophagy in biomedicine are nonnegligible and require our particular concerns. MAIN BODY We collected research on the toxic effects related to NM-mediated autophagy both in vivo and in vitro. Generally, NMs can be delivered into animal models through different administration routes, or internalized by cells through different uptake pathways, exerting varying degrees of damage in tissues, organs, cells, and organelles, eventually being deposited in or excreted from the body. In addition, other biological effects of NMs, such as oxidative stress, inflammation, necroptosis, pyroptosis, and ferroptosis, have been associated with autophagy and cooperate to regulate body activities. We therefore highlight that NM-mediated autophagy serves as a double-edged sword, which could be utilized in the treatment of certain diseases related to autophagy dysfunction, such as cancer, neurodegenerative disease, and cardiovascular disease. Challenges and suggestions for further investigations of NM-mediated autophagy are proposed with the purpose to improve their biosafety evaluation and facilitate their wide application. Databases such as PubMed and Web of Science were utilized to search for relevant literature, which included all published, Epub ahead of print, in-process, and non-indexed citations. CONCLUSION In this review, we focus on the dual effect of NM-mediated autophagy in the biomedical field. It has become a trend to use the benefits of NM-mediated autophagy to treat clinical diseases such as cancer and neurodegenerative diseases. Understanding the regulatory mechanism of NM-mediated autophagy in biomedicine is also helpful for reducing the toxic effects of NMs as much as possible.
Collapse
Affiliation(s)
- Xiaoli Feng
- Stomatological Hospital, Southern Medical University, 366 South Jiangnan Road, Guangzhou, 510280, China
| | - Yaqing Zhang
- Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Street, Guangzhou, 510515, China
| | - Chao Zhang
- Orthodontic Department, Stomatological Hospital, Southern Medical University, 366 South Jiangnan Road, Guangzhou, 510280, China
| | - Xuan Lai
- Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Street, Guangzhou, 510515, China
| | - Yanli Zhang
- Stomatological Hospital, Southern Medical University, 366 South Jiangnan Road, Guangzhou, 510280, China
| | - Junrong Wu
- Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Street, Guangzhou, 510515, China
| | - Chen Hu
- Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Street, Guangzhou, 510515, China
| | - Longquan Shao
- Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Street, Guangzhou, 510515, China.
| |
Collapse
|
21
|
Pyrogenic and Precipitated Amorphous Silica Nanoparticles Differentially Affect Cell Responses to LPS in Human Macrophages. NANOMATERIALS 2020; 10:nano10071395. [PMID: 32708373 PMCID: PMC7407657 DOI: 10.3390/nano10071395] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/09/2020] [Accepted: 07/14/2020] [Indexed: 12/23/2022]
Abstract
Previous work has demonstrated that precipitated (NM-200) and pyrogenic (NM-203) Amorphous Silica Nanoparticles (ASNPs) elicit the inflammatory activation of murine macrophages, with more pronounced effects observed with NM-203. Here, we compare the effects of low doses of NM-200 and NM-203 on human macrophage-like THP-1 cells, assessing how the pre-exposure to these nanomaterials affects the cell response to lipopolysaccharide (LPS). Cell viability was affected by NM-203, but not by NM-200, and only in the presence of LPS. While NM-203 stimulated mTORC1, neither ASNPs activated NFκB or the transcription of its target genes PTGS2 and IL1B. NM-200 and NM-203 caused a block of the autophagic flux and inhibited the LPS-dependent increase of Glutamine Synthetase (GS) expression. Both ASNPs suppressed the activation of caspase-1, delaying the LPS-dependent secretion of IL-1β. Thus, ASNPs modulate several important pathways in human macrophages, altering their response to LPS. NM-203 had larger effects on autophagy, mTORC1 activity and GS expression than NM-200, confirming the higher biological activity of pyrogenic ASNPs when compared with precipitated ASNPs.
Collapse
|
22
|
Wu Y, Jin Y, Sun T, Zhu P, Li J, Zhang Q, Wang X, Jiang J, Chen G, Zhao X. p62/SQSTM1 accumulation due to degradation inhibition and transcriptional activation plays a critical role in silica nanoparticle-induced airway inflammation via NF-κB activation. J Nanobiotechnology 2020; 18:77. [PMID: 32429946 PMCID: PMC7236097 DOI: 10.1186/s12951-020-00634-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Most nanoparticles (NPs) reportedly block autophagic flux, thereby upregulating p62/SQSTM1 through degradation inhibition. p62 also acts as a multifunctional scaffold protein with multiple domains, and is involved in various cellular processes. However, the autophagy substrate-independent role of p62 and its regulation at the transcriptional level upon NPs exposure remain unclear. RESULTS In this work, we exposed BEAS-2b cells and mice to silica nanoparticles (SiNPs), and found that SiNPs increased p62 protein levels in vivo and vitro. Then, we further explored the role and mechanism of SiNPs-stimulated p62 in vitro, and found that p62 degradation was inhibited due to autophagic flux blockade. Mechanistically, SiNPs blocked autophagic flux through impairment of lysosomal capacity rather than defective autophagosome fusion with lysosomes. Moreover, SiNPs stimulated translocation of NF-E2-related factor 2 (Nrf2) to the nucleus from the cytoplasm, which upregulated p62 transcriptional activation through direct binding of Nrf2 to the p62 promoter. Nrf2 siRNA dramatically reduced both the mRNA and protein levels of p62. These two mechanisms led to p62 protein accumulation, thus increasing interleukin (IL)-1 and IL-6 expression. SiNPs activated nuclear factor kappa B (NF-κB), and this effect could be alleviated by p62 knockdown. CONCLUSION SiNPs caused accumulation of p62 through both pre- and post-translational mechanisms, resulting in airway inflammation. These findings improve our understanding of SiNP-induced pulmonary damage and the molecular targets available to mitigate it.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nangtong University, Nantong, 226019, China
| | - Yang Jin
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nangtong University, Nantong, 226019, China
| | - Tianyu Sun
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nangtong University, Nantong, 226019, China
| | - Piaoyu Zhu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nangtong University, Nantong, 226019, China
| | - Jinlong Li
- School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Qinglin Zhang
- Departments of Gastroenterology, Affiliated to Wuxi People's Hospital, Nanjing Medical University, Wuxi, 214023, China
| | - Xiaoke Wang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nangtong University, Nantong, 226019, China
| | - Junkang Jiang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nangtong University, Nantong, 226019, China
| | - Gang Chen
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nangtong University, Nantong, 226019, China.
| | - Xinyuan Zhao
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nangtong University, Nantong, 226019, China.
| |
Collapse
|
23
|
Wu H, Wang X, Liang H, Zheng J, Huang S, Zhang D. Enhanced efficacy of propranolol therapy for infantile hemangiomas based on a mesoporous silica nanoplatform through mediating autophagy dysfunction. Acta Biomater 2020; 107:272-285. [PMID: 32145394 DOI: 10.1016/j.actbio.2020.02.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/13/2020] [Accepted: 02/21/2020] [Indexed: 12/17/2022]
Abstract
Infantile hemangioma is one of the most common vascular tumors, which might result in morbidity and mortality without timely intervention. Propranolol is currently the first-line therapy for hemangiomas, but its potential side effects and high frequency of administration make it urgent to develop a suitable drug delivery system for propranolol. In the present study, we formulated a propranolol delivery system based on mesoporous silica nanoparticles (PRN@MSN) and investigated the interplay between autophagic activities mediated by nanoparticles and improved therapeutic efficacy of PRN@MSN. The results showed that PRN@MSN nanoparticles exhibited higher cytotoxicity compared with free propranolol in vitro and in vivo, which could induce excessive autophagosome accumulation through increased autophagosome formation and impaired autophagic degradation. Inhibition of autophagy in the early stage could attenuate the cytotoxicity of PRN@MSN. ROS generation was essential for nanoparticle-mediated autophagy and cytotoxicity, and PRN@MSN-induced autophagy dysfunction could enhance endoplasmic reticulum (ER) stress in hemangioma stem cells. Our study revealed a promising PRN delivery system based on a mesoporous silica nanoplatform that could induce autophagy dysfunction with excessive autophagosome accumulation to promote the therapeutic efficacy of PRN therapy. PRN@MSN drug delivery system combined with autophagy modulation may act as a promising treatment pattern in the treatment of hemangiomas.
Collapse
Affiliation(s)
- Haiwei Wu
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250012, China; Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250012, China
| | - Xuan Wang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250012, China; Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250012, China
| | - Hao Liang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250012, China; Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250012, China
| | - Jiawei Zheng
- Department of Oral and Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Shengyun Huang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250012, China; Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250012, China.
| | - Dongsheng Zhang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250012, China; Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
24
|
Li Y, Duan J, Chai X, Yang M, Wang J, Chen R, Sun Z. Microarray-assisted size-effect study of amorphous silica nanoparticles on human bronchial epithelial cells. NANOSCALE 2019; 11:22907-22923. [PMID: 31763651 DOI: 10.1039/c9nr07350g] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Amorphous silica nanoparticles (SiNPs) are not only abundant in nature, but also the second largest engineering nanomaterials in terms of annual output. Respiratory exposure is the main route for SiNPs to enter the human body. A large number of studies have focused on the respiratory toxicity of SiNPs and demonstrated that SiNPs could induce pulmonary tissue damage, inflammation, fibrosis, and even the malignant transformation of bronchial epithelial cells, while the size-dependent toxicity of SiNPs and their underlying biological mechanisms remain unclear. In this regard, a transcriptomics study would be conductive to gaining a better understanding of the toxic mechanism. In the present study, microarray analysis was performed to investigate the genome-wide transcriptional alteration induced by different sizes of SiNPs in human primary bronchial epithelial cells (BEAS-2B). To determine the effect of the particle size on the toxicity, nanoparticles of two sizes (41 nm and 61 nm) and submicron particles of one size (206 nm) were introduced. The bioinformatics analysis results indicated that: (1) the number of differentially expressed genes in the three SiNP-treated groups increased with the particle size decreasing; (2) the genes involved in the immune and inflammatory response, gene expression, signal transduction, endoplasmic reticulum stress, oxidative stress, cell metabolism, and cell proliferation were gradually upregulated with the particle size decreasing, while the genes related to the morphological development of the respiratory system were gradually downregulated with the particle size decreasing; (3) the modes of action of the two nanoparticles overlapped with each other to some degree, and there existed many different modes compared to those from the submicron particles; (4) both the silica nanoparticles affected the pathways associated with the cell entry of silica nanoparticles, autophagy and lysosomal dysfunction, endoplasmic reticulum stress, inflammatory response, DNA damage, and gene expression, as well as apoptotic resistance and cancer. To the best of our knowledge, this is the first study that has reported the alteration trend of gene expression profiles with the change in silica particle size. Our study provides a great deal of information on the toxic mechanisms underlying the respiratory toxicity induced by SiNPs, and can also serve as an experimental basis for the toxicity and safety evaluation of silica nanoparticles.
Collapse
Affiliation(s)
- Yang Li
- School of Public Health, Beijing, 100069, China and Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| | - Junchao Duan
- School of Public Health, Beijing, 100069, China and Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| | - Xiangyuan Chai
- Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Man Yang
- School of Public Health, Beijing, 100069, China and Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| | - Ji Wang
- School of Public Health, Beijing, 100069, China and Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| | - Rui Chen
- School of Public Health, Beijing, 100069, China and Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| | - Zhiwei Sun
- School of Public Health, Beijing, 100069, China and Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
25
|
Kim TH, Kang MS, Mandakhbayar N, El-Fiqi A, Kim HW. Anti-inflammatory actions of folate-functionalized bioactive ion-releasing nanoparticles imply drug-free nanotherapy of inflamed tissues. Biomaterials 2019; 207:23-38. [DOI: 10.1016/j.biomaterials.2019.03.034] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 03/21/2019] [Accepted: 03/22/2019] [Indexed: 01/04/2023]
|
26
|
Hočevar S, Milošević A, Rodriguez-Lorenzo L, Ackermann-Hirschi L, Mottas I, Petri-Fink A, Rothen-Rutishauser B, Bourquin C, Clift MJD. Polymer-Coated Gold Nanospheres Do Not Impair the Innate Immune Function of Human B Lymphocytes in Vitro. ACS NANO 2019; 13:6790-6800. [PMID: 31117377 DOI: 10.1021/acsnano.9b01492] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Gold nanoparticles (GNPs) are intended for use within a variety of biomedical applications due to their physicochemical properties. Although, in general, biocompatibility of GNPs with immune cells such as macrophages and dendritic cells is well established, the impact of GNPs on B lymphocyte immune function remains to be determined. Since B lymphocytes play an important role in health and disease, the suitability of GNPs as a B cell-targeting tool is of high relevance. Thus, we provide information on the interactions of GNPs with B lymphocytes. Herein, we exposed freshly isolated human B lymphocytes to a set of well-characterized and biomedically relevant GNPs with distinct surface (polyethylene glycol (PEG), PEG/poly(vinyl alcohol) (PEG/PVA)) and shape (spheres, rods) characteristics. Polymer-coated GNPs poorly interacted with B lymphocytes, in contrast to uncoated GNPs. Importantly, none of the GNPs significantly affected cell viability, even at the highest concentration of 20 μg/mL over a 24 h suspension exposure period. Furthermore, none of the nanosphere formulations affected the expression of activation markers (CD69, CD86, MHC II) of the naive B lymphocytes, nor did they cause an increase in the secretion of pro-inflammatory cytokines ( i.e. , IL-6, IL-1β). However, the absence of polymer coating on the sphere GNPs and the rod shape caused a decrease in IL-6 cytokine production by activated B lymphocytes, suggesting a functional impairment. With these findings, the present study contributes imperative knowledge toward the safe-by-design approaches being conducted to benefit the development of nanomaterials, specifically those as theranostic tools.
Collapse
Affiliation(s)
- Sandra Hočevar
- BioNanomaterials , Adolphe Merkle Institute, University of Fribourg , 1700 Fribourg , Switzerland
- School of Pharmaceutical Sciences , University of Geneva, University of Lausanne , 1211 Geneva , Switzerland
| | - Ana Milošević
- BioNanomaterials , Adolphe Merkle Institute, University of Fribourg , 1700 Fribourg , Switzerland
| | - Laura Rodriguez-Lorenzo
- BioNanomaterials , Adolphe Merkle Institute, University of Fribourg , 1700 Fribourg , Switzerland
| | | | - Ines Mottas
- School of Pharmaceutical Sciences , University of Geneva, University of Lausanne , 1211 Geneva , Switzerland
- Chair of Pharmacology, Faculty of Science and Medicine , University of Fribourg , 1700 Fribourg , Switzerland
| | - Alke Petri-Fink
- BioNanomaterials , Adolphe Merkle Institute, University of Fribourg , 1700 Fribourg , Switzerland
| | | | - Carole Bourquin
- School of Pharmaceutical Sciences , University of Geneva, University of Lausanne , 1211 Geneva , Switzerland
- Chair of Pharmacology, Faculty of Science and Medicine , University of Fribourg , 1700 Fribourg , Switzerland
- Faculty of Medicine , University of Geneva , Rue Michel-Servet 1 , 1211 Geneva , Switzerland
| | - Martin James David Clift
- BioNanomaterials , Adolphe Merkle Institute, University of Fribourg , 1700 Fribourg , Switzerland
- In Vitro Toxicology Group , Swansea University Medical School , Wales SA2 8PP , U.K
| |
Collapse
|
27
|
Riediker M, Zink D, Kreyling W, Oberdörster G, Elder A, Graham U, Lynch I, Duschl A, Ichihara G, Ichihara S, Kobayashi T, Hisanaga N, Umezawa M, Cheng TJ, Handy R, Gulumian M, Tinkle S, Cassee F. Particle toxicology and health - where are we? Part Fibre Toxicol 2019; 16:19. [PMID: 31014371 PMCID: PMC6480662 DOI: 10.1186/s12989-019-0302-8] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 04/08/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Particles and fibres affect human health as a function of their properties such as chemical composition, size and shape but also depending on complex interactions in an organism that occur at various levels between particle uptake and target organ responses. While particulate pollution is one of the leading contributors to the global burden of disease, particles are also increasingly used for medical purposes. Over the past decades we have gained considerable experience in how particle properties and particle-bio interactions are linked to human health. This insight is useful for improved risk management in the case of unwanted health effects but also for developing novel medical therapies. The concepts that help us better understand particles' and fibres' risks include the fate of particles in the body; exposure, dosimetry and dose-metrics and the 5 Bs: bioavailability, biopersistence, bioprocessing, biomodification and bioclearance of (nano)particles. This includes the role of the biomolecule corona, immunity and systemic responses, non-specific effects in the lungs and other body parts, particle effects and the developing body, and the link from the natural environment to human health. The importance of these different concepts for the human health risk depends not only on the properties of the particles and fibres, but is also strongly influenced by production, use and disposal scenarios. CONCLUSIONS Lessons learned from the past can prove helpful for the future of the field, notably for understanding novel particles and fibres and for defining appropriate risk management and governance approaches.
Collapse
Affiliation(s)
- Michael Riediker
- Swiss Centre for Occupational and Environmental Health (SCOEH), Binzhofstrasse 87, CH-8404 Winterthur, Switzerland
| | - Daniele Zink
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Wolfgang Kreyling
- Institute of Epidemiology, Helmholtz Center Munich – German Research Center for Environmental Health, Neuherberg, Munich Germany
| | - Günter Oberdörster
- Department of Environmental Medicine, University of Rochester, Rochester, NY USA
| | - Alison Elder
- Department of Environmental Medicine, University of Rochester, Rochester, NY USA
| | | | - Iseult Lynch
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Albert Duschl
- Department of Biosciences, Allergy Cancer BioNano Research Centre, University of Salzburg, Salzburg, Austria
| | | | | | | | | | | | | | - Richard Handy
- School of Biological Sciences, Plymouth University, Plymouth, UK
| | - Mary Gulumian
- National Institute for Occupational Health and Haematology and Molecular Medicine, University of the Witwatersrand, Johannesburg, South Africa
| | - Sally Tinkle
- Science and Technology Policy Institute, Washington, DC USA
| | - Flemming Cassee
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
- Institute for Risk Assessment Studies (IRAS), Utrrecht University, Utrecht, The Netherlands
| |
Collapse
|
28
|
Su Y, Lu J, Chen X, Liang C, Luo P, Qin C, Zhang J. Rapamycin Alleviates Hormone Imbalance-Induced Chronic Nonbacterial Inflammation in Rat Prostate Through Activating Autophagy via the mTOR/ULK1/ATG13 Signaling Pathway. Inflammation 2018; 41:1384-1395. [PMID: 29675586 DOI: 10.1007/s10753-018-0786-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chronic prostatitis (CP) is a clinically common disease with high morbidity. It affects the patients' quality of life (QoL) as well as physical and mental health seriously due to the recurring symptoms of lower urinary tract and genitalia. As the opinions about the etiology of CP are still not uniform, it is very difficult to be treated or even cured. Autophagy is a highly conserved physiological function which is widely found in eukaryotic cells. In general, cells maintain a certain level of autophagy under physiological conditions, and the basal level of autophagy can be regulated by a variety of autophagy-related genes under stress such as hunger, infection, trauma, and other circumstances. Therefore, the main purpose of this study is to investigate the role of autophagy in chronic nonbacterial prostatitis (CNP, also called CP). In this paper, we established the CNP model via hypodermic injection of 17β-estradiol and subsequently abdominal rapamycin (a common autophagy inducer) treatment based on castrated rats. Then, the expression of nuclear factor-κB (NF-κB), interleukin-1β (IL-1β), and autophagy-related markers as well as autophagosome formation in prostate tissues, peripheral blood mononuclear cells (PBMCs), and serum of rats were evaluated respectively. In addition to some histological changes in the prostate tissues, we found the levels of NF-κB and IL-1β were significantly increased in the model group, along with significantly suppressed autophagy, whereas rapamycin could reverse these effects which involved in the mTOR/ULK1/ATG13 signaling pathway. In conclusion, our results suggested that rapamycin could ameliorate hormone imbalance-induced CNP by activating autophagy.
Collapse
Affiliation(s)
- Yang Su
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jingxiao Lu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xianguo Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Pengcheng Luo
- Huangshi Central Hospital, Hubei Polytechnic University, Huangshi, 435000, China
| | - Cong Qin
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jie Zhang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China. .,Huangshi Central Hospital, Hubei Polytechnic University, Huangshi, 435000, China.
| |
Collapse
|
29
|
Wei F, Duan Y. Crosstalk between Autophagy and Nanomaterials: Internalization, Activation, Termination. ACTA ACUST UNITED AC 2018; 3:e1800259. [PMID: 32627344 DOI: 10.1002/adbi.201800259] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/02/2018] [Indexed: 12/12/2022]
Abstract
Nanomaterials (NMs) are comprehensively applied in biomedicine due to their unique physical and chemical properties. Autophagy, as an evolutionarily conserved cellular quality control process, is closely associated with the effect of NMs on cells. In this review, the recent advances in NM-induced/inhibited autophagy (NM-phagy) are summarized, with an aim to present a comprehensive description of the mechanisms of NM-phagy from the perspective of internalization, activation, and termination, thereby bridging autophagy and nanomaterials. Several possible mechanisms are extensively reviewed including the endocytosis pathway of NMs and the related cross components (clathrin and adaptor protein 2 (AP-2), adenosine diphosphate (ADP)-ribosylation factor 6 (Arf6), Rab, UV radiation resistance associated gene (UVRAG)), three main stress mechanisms (oxidative stress, damaged organelles stress, and toxicity stress), and several signal pathway-related molecules. The mechanistic insight is beneficial to understand the autophagic response to NMs or NMs' regulation of autophagy. The challenges currently encountered and research trend in the field of NM-phagy are also highlighted. It is hoped that the NM-phagy discussion in this review with the focus on the mechanistic aspects may serve as a guideline for future research in this field.
Collapse
Affiliation(s)
- Fujing Wei
- Research Center of Analytical Instrumentation, Key Laboratory of Bio-resource and Eco-enviroment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, P. R. China
| | - Yixiang Duan
- Research Center of Analytical Instrumentation, Key Laboratory of Bio-resource and Eco-enviroment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, P. R. China
| |
Collapse
|
30
|
Chen L, Liu J, Zhang Y, Zhang G, Kang Y, Chen A, Feng X, Shao L. The toxicity of silica nanoparticles to the immune system. Nanomedicine (Lond) 2018; 13:1939-1962. [PMID: 30152253 DOI: 10.2217/nnm-2018-0076] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Silicon-based materials and their oxides are widely used in drug delivery, dietary supplements, implants and dental fillers. Silica nanoparticles (SiNPs) interact with immunocompetent cells and induce immunotoxicity. However, the toxic effects of SiNPs on the immune system have been inadequately reviewed. The toxicity of SiNPs to the immune system depends on their physicochemical properties and the cell type. Assessments of immunotoxicity include determining cell dysfunctions, cytotoxicity and genotoxicity. This review focuses on the immunotoxicity of SiNPs and investigates the underlying mechanisms. The main mechanisms were proinflammatory responses, oxidative stress and autophagy. Considering the toxicity of SiNPs, surface and shape modifications may mitigate the toxic effects of SiNPs, providing a new way to produce these nanomaterials with less toxic impaction.
Collapse
Affiliation(s)
- Liangjiao Chen
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou 510140, PR China
| | - Jia Liu
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Yanli Zhang
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Guilan Zhang
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Yiyuan Kang
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Aijie Chen
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Xiaoli Feng
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Longquan Shao
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| |
Collapse
|
31
|
Wang J, Li Y, Duan J, Yang M, Yu Y, Feng L, Yang X, Zhou X, Zhao Z, Sun Z. Silica nanoparticles induce autophagosome accumulation via activation of the EIF2AK3 and ATF6 UPR pathways in hepatocytes. Autophagy 2018; 14:1185-1200. [PMID: 29940794 DOI: 10.1080/15548627.2018.1458174] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Autophagy dysfunction is a potential toxic effect of nanoparticles. Previous studies have indicated that silica nanoparticles (SiNPs) induce macroautophagy/autophagy dysfunction, while the precise mechanisms remain uncertain. Hence, the present study investigated the molecular mechanisms by which SiNPs enhanced autophagosome synthesis, which then contributed to autophagy dysfunction. First, the effects of SiNPs on autophagy and autophagic flux were verified using transmission electron microscopy, laser scanning confocal microscopy, and western blot assays. Then, the activation of endoplasmic reticular (ER) stress was validated to be through the EIF2AK3 and ATF6 UPR pathways but not the ERN1-XBP1 pathway, along with the upregulation of downstream ATF4 and DDIT3. Thereafter, the ER stress inhibitor 4-phenylbutyrate (4-PBA) was used to verify that SiNP-induced autophagy could be influenced by ER stress. Furthermore, specialized lentiviral shRNA were employed to determine that autophagy was induced via specific activation of the EIF2AK3 and ATF6 UPR pathways. Finally, the 2 autophagic genes LC3B and ATG12 were found to be transcriptionally upregulated by downstream ATF4 and DDIT3 in ER stress, which contributed to the SiNP-enhanced autophagosome synthesis. Taken together, these data suggest that SiNPs induced autophagosome accumulation via the activation of the EIF2AK3 and ATF6 UPR pathways in hepatocytes, which offers a new insight into detailed molecular mechanisms underlying SiNP-induced autophagy dysfunction, and specifically how UPR pathways regulate key autophagic genes. This work provides novel evidence for the study of toxic effects and risk assessment of SiNPs.
Collapse
Affiliation(s)
- Ji Wang
- a Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Yang Li
- a Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Junchao Duan
- a Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Man Yang
- a Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Yang Yu
- a Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Lin Feng
- a Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Xiaozhe Yang
- a Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Xianqing Zhou
- a Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| | - Zhendong Zhao
- c MOH Key Laboratory of Systems Biology of Pathogens , Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College , Beijing , China
| | - Zhiwei Sun
- a Department of Toxicology and Sanitary Chemistry , School of Public Health, Capital Medical University , Beijing , P.R. China.,b Beijing Key Laboratory of Environmental Toxicology , Capital Medical University , Beijing , P.R. China
| |
Collapse
|
32
|
Silibinin-induced autophagy mediated by PPARα-sirt1-AMPK pathway participated in the regulation of type I collagen-enhanced migration in murine 3T3-L1 preadipocytes. Mol Cell Biochem 2018; 450:1-23. [DOI: 10.1007/s11010-018-3368-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 05/17/2018] [Indexed: 12/21/2022]
|
33
|
Huang YJ, Hung KC, Hung HS, Hsu SH. Modulation of Macrophage Phenotype by Biodegradable Polyurethane Nanoparticles: Possible Relation between Macrophage Polarization and Immune Response of Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2018; 10:19436-19448. [PMID: 29775050 DOI: 10.1021/acsami.8b04718] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Nanomaterials with surface functionalized by different chemical groups can either provoke or attenuate the immune responses of the nanomaterials, which is critical to their biomedical efficacies. In this study, we demonstrate that synthetic waterborne polyurethane nanoparticles (PU NPs) can inhibit the macrophage polarization toward the M1 phenotype but not M2 phenotype. The surface-functionalized PU NPs decrease the secretion levels of proinflammatory cytokines (TNF-α and IL-1β) for M1 macrophages. Specifically, PU NPs with carboxyl groups on the surface exhibit a greater extent of inhibition on M1 polarization than those with amine groups. These water-suspended PU NPs reduce the nuclear factor-κB (NF-κB) activation and suppress the subsequent NLR family pyrin domain containing 3 (NLRP3) inflammasome signals. Furthermore, the dried PU films assembled from PU NPs have a similar effect on macrophage polarization and present a smaller shifting foreign body reaction (FBR) in vivo than the conventional poly(l-lactic acid). Taken together, the biodegradable waterborne PU NPs demonstrate surface-dependent immunosuppressive properties and macrophage polarization effects. The findings suggest potential therapeutic applications of PU NPs in anti-inflammation and macrophage-related disorders and propose a mechanism for the low FBR observed for biodegradable PU materials.
Collapse
Affiliation(s)
- Yen-Jang Huang
- Institute of Polymer Science and Engineering , National Taiwan University , Taipei , Taiwan 106 , R.O.C
| | - Kun-Che Hung
- Institute of Polymer Science and Engineering , National Taiwan University , Taipei , Taiwan 106 , R.O.C
| | - Huey-Shan Hung
- Translational Medicine Research , China Medical University Hospital , Taichung , Taiwan 404 , R.O.C
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering , National Taiwan University , Taipei , Taiwan 106 , R.O.C
- Institute of Cellular and System Medicine , National Health Research Institutes , Miaoli County , Taiwan 350 , R.O.C
| |
Collapse
|
34
|
Wang Y, Zhao Z, Wei F, Luo Z, Duan Y. Combining autophagy-inducing peptides and brefeldin A delivered by perinuclear-localized mesoporous silica nanoparticles: a manipulation strategy for ER-phagy. NANOSCALE 2018; 10:8796-8805. [PMID: 29713715 DOI: 10.1039/c8nr00872h] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Autophagic degradation of the endoplasmic reticulum (ER-phagy) has been found to play a critical role in human sensory neuropathy. So far, however, specific and efficient intervention means for ER-phagy remain unexplored. Herein, brefeldin A (BFA), a blocking agent on protein transport between the ER and Golgi, was screened from ER stress inducers. BFA was then delivered to the perinuclear area co-localized with the ER by a mesoporous silica nanoparticle-based drug-carrier functionalized with autophagy-inducing peptides of TAT-beclin 1 (MSNs-BFA), to evoke a perturbation of ER-phagy. The molecular mechanism of ER-phagy regulated by BFA was explored by biochemical evaluation including time-lapse live-cell fluorescence imaging. We found that MSNs-BFA treatment caused a lower mRNA/protein expression level of FAM134b even under a compensation of autophagic flux in U2OS cells, and resulted in ER-expansion. The fragmentation of the ER was blocked as a response to ER stress mediated by inactivation of the AKT/TSC/mTOR pathway. Our work developed an efficient external manipulation strategy to regulate ER-phagy and may contribute to the therapeutic application of autophagy-related major human diseases.
Collapse
Affiliation(s)
- Yimin Wang
- Research Center of Analytical Instrumentation, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610064, Sichuan, PR China.
| | | | | | | | | |
Collapse
|
35
|
Lin XT, Zheng XB, Fan DJ, Yao QQ, Hu JC, Lian L, Wu XJ, Lan P, He XS. MicroRNA-143 Targets ATG2B to Inhibit Autophagy and Increase Inflammatory Responses in Crohn's Disease. Inflamm Bowel Dis 2018; 24:781-791. [PMID: 29562274 DOI: 10.1093/ibd/izx075] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Dysfunctional autophagy is recognized as a contributing factor in many chronic inflammatory diseases, including Crohn's disease (CD). Genetic analyses have found that microRNA (miRNA) levels are altered in the intestinal tissues of CD patients. METHODS The Sequencing Alternative Poly-Adenylation Sites (SAPAS) method was used to compare the 3' end of the total mRNA sequence of 3 surgical specimens of CD patients (including inflamed tissues and corresponding noninflamed tissues in each case). The levels of autophagy-related 2B (ATG2B), LC3, and miR-143 were compared between inflamed tissues and noninflamed tissues using immunoblot and quantitative reverse transcription polymerase chain reaction. Luciferase assays were used to verify the interactions between miR-143 and ATG2B. Autophagy was measured by immunoblot analyses of LC3 and transmission electron microscopy. Inflammatory cytokines and IκBα were analyzed to evaluate the effect of miR-143 on inflammatory response. RESULTS The tandem repeat 3'-UTR of ATG2B was longer in inflamed tissues than in corresponding noninflamed tissues and contained an miR-143 target site. miR-143 expression was elevated, whereas ATG2B and LC3-II were downregulated in inflamed tissues. The direct interaction between miR-143 and ATG2B was verified by a 3'-UTR dual-luciferase reporter assay. Constitutive expression of miR-143 or depletion of ATG2B in cultured intestinal epithelial cells inhibited autophagy, reduced IκBα levels, and increased inflammatory responses. CONCLUSIONS miR-143 may induce bowel inflammation by regulating ATG2B and autophagy, suggesting that miR-143 might play a critical role in the development of CD. Therefore, miR-143 could be a promising novel target for gene therapy in CD patients.
Collapse
Affiliation(s)
- Xu-Tao Lin
- Department of Gastrointestinal Endoscopy, Guangzhou, Guangdong, China.,Department of Colorectal Surgery, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao-Bin Zheng
- Department of Colorectal Surgery, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - De-Jun Fan
- Department of Gastrointestinal Endoscopy, Guangzhou, Guangdong, China.,Department of Colorectal Surgery, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qiu-Qiong Yao
- Department of Colorectal Surgery, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jian-Cong Hu
- Department of Colorectal Surgery, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lei Lian
- Department of Colorectal Surgery, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao-Jian Wu
- Department of Colorectal Surgery, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ping Lan
- Department of Colorectal Surgery, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao-Sheng He
- Department of Colorectal Surgery, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
36
|
Dalzon B, Aude-Garcia C, Collin-Faure V, Diemer H, Béal D, Dussert F, Fenel D, Schoehn G, Cianférani S, Carrière M, Rabilloud T. Differential proteomics highlights macrophage-specific responses to amorphous silica nanoparticles. NANOSCALE 2017; 9:9641-9658. [PMID: 28671223 DOI: 10.1039/c7nr02140b] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
The technological and economic benefits of engineered nanomaterials may be offset by their adverse effects on living organisms. One of the highly produced nanomaterials under such scrutiny is amorphous silica nanoparticles, which are known to have an appreciable, although reversible, inflammatory potential. This is due to their selective toxicity toward macrophages, and it is thus important to study the cellular responses of this cell type to silica nanoparticles to better understand the direct or indirect adverse effects of nanosilica. We have here studied the responses of the RAW264.7 murine macrophage cells and of the control MPC11 plasma cells to subtoxic concentrations of nanosilica, using a combination of proteomic and targeted approaches. This allowed us to document alterations in the cellular cytoskeleton, in the phagocytic capacity of the cells as well as their ability to respond to bacterial stimuli. More surprisingly, silica nanoparticles also induce a greater sensitivity of macrophages to DNA alkylating agents, such as styrene oxide, even at doses which do not induce any appreciable cell death.
Collapse
Affiliation(s)
- Bastien Dalzon
- Laboratory of Chemistry and Biology of Metals, UMR 5249, Univ. Grenoble Alpes, CNRS, CEA, Grenoble, France.
| | - Catherine Aude-Garcia
- Laboratory of Chemistry and Biology of Metals, UMR 5249, Univ. Grenoble Alpes, CNRS, CEA, Grenoble, France.
| | - Véronique Collin-Faure
- Laboratory of Chemistry and Biology of Metals, UMR 5249, Univ. Grenoble Alpes, CNRS, CEA, Grenoble, France.
| | - Hélène Diemer
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - David Béal
- Chimie Interface Biologie pour l'Environnement, la Santé et la Toxicologie (CIBEST), UMR 5819, Univ. Grenoble Alpes, CEA, CNRS, INAC, SyMMES, F-38000 Grenoble, France
| | - Fanny Dussert
- Chimie Interface Biologie pour l'Environnement, la Santé et la Toxicologie (CIBEST), UMR 5819, Univ. Grenoble Alpes, CEA, CNRS, INAC, SyMMES, F-38000 Grenoble, France
| | - Daphna Fenel
- Institut de Biologie Structurale Jean-Pierre Ebel, UMR5075, Univ. Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Guy Schoehn
- Institut de Biologie Structurale Jean-Pierre Ebel, UMR5075, Univ. Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Marie Carrière
- Laboratoire de Spectrométrie de Masse BioOrganique, Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Thierry Rabilloud
- Laboratory of Chemistry and Biology of Metals, UMR 5249, Univ. Grenoble Alpes, CNRS, CEA, Grenoble, France.
| |
Collapse
|
37
|
Chou CC, Chen W, Hung Y, Mou CY. Molecular Elucidation of Biological Response to Mesoporous Silica Nanoparticles in Vitro and in Vivo. ACS APPLIED MATERIALS & INTERFACES 2017; 9:22235-22251. [PMID: 28608695 DOI: 10.1021/acsami.7b05359] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Biomedical applications of mesoporous silica nanoparticles (MSNs) require efficient cellular uptake and low toxicity. The purpose of this study is to investigate the cellular uptake and toxicity of MSNs with different sizes and charges (50, 100, and 250 nm with a positive surface charge and 100 nm with a negative surface charge) exposed to human monocyte-derived macrophages, lung epithelium BEAS-2B cells, and mice using genome-wide gene expression analysis and cellular/animal-level end point tests. We found that MSNs can be taken up into cells through endocytosis in a charge- and size-dependent manner, with positively charged and larger MSNs being more easily taken up into the cells by recruiting more types of endocytotic pathways for more cellular uptake. Moreover, the cytotoxicity of MSNs could be correlated with the amount of MSNs taken up by cells, which positively correlates to the particle size and dosage. Therefore, only positively charged and larger MSNs (≥100 nm) during higher treatment doses (≥500 μg mL-1) resulted in a sufficient accumulation of internalized MSNs in cells to induce significant release of reactive oxygen species (ROS) and oxidative stress, inflammatory gene upregulation through NF-κB and AP-1, and eventually autophagy-mediated necrotic cell death. Furthermore, genome-wide gene expression analysis could reflect the above in vitro cellular damages and corresponding in vivo injuries in mice, indicating that specific gene expression footprints may be used for assessing the safety of nanoparticles. The present finding provides some insights into the rational design of effective MSN-based drug/gene delivery systems and biomedical applications.
Collapse
Affiliation(s)
- Cheng-Chung Chou
- Department of Life Science and Institute of Molecular Biology, National Chung Cheng University , Chia-Yi, Taiwan 62102, ROC
| | - Wei Chen
- Department of Chemistry, National Taiwan University , Taipei, Taiwan 10617, ROC
| | - Yann Hung
- Department of Chemistry, National Taiwan University , Taipei, Taiwan 10617, ROC
| | - Chung-Yuan Mou
- Department of Chemistry, National Taiwan University , Taipei, Taiwan 10617, ROC
| |
Collapse
|
38
|
Wei F, Wang Y, Luo Z, Li Y, Duan Y. New findings of silica nanoparticles induced ER autophagy in human colon cancer cell. Sci Rep 2017; 7:42591. [PMID: 28195184 PMCID: PMC5307363 DOI: 10.1038/srep42591] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 01/10/2017] [Indexed: 01/07/2023] Open
Abstract
Nanoparticle-induced autophagy has been extensively studied, however, real time information about the endoplasmic reticulum involved autophagic process (ER autophagy) induced by nanomaterials remains unknown. In this work, silica nanoparticles (SNPs) were synthesized with characteristics of low toxicity, good biocompatibility and excellent water dispersibility to treat cells. Results show that either low concentration (10 μg/mL) or high concentration (200 μg/mL) of SNPs could increase the quantity of processing from microtubule-associated protein 1-light chain 3-I (LC3-I) to the other variant of LC3 (LC3-II). Interestingly, the level of autophagy induced by the SNPs is associated with the treated time but not the concentrations of SNPs. Importantly, for the first time, SNP accumulation in ER was discovered through co-localization analysis, which incurs ER autophagy. These new findings about SNPs-induced ER autophagy could open an effective way for securely designing silica-based nanoparticles and enable us to know more about ER autophagy.
Collapse
Affiliation(s)
- Fujing Wei
- Research Center of Analytical Instrumentation, Key Laboratory of Bio-resource and Eco-environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, PR China
| | - Yimin Wang
- Research Center of Analytical Instrumentation, Key Laboratory of Bio-resource and Eco-environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, PR China
| | - Zewei Luo
- Research Center of Analytical Instrumentation, Key Laboratory of Bio-resource and Eco-environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, PR China
| | - Yu Li
- Research Center of Analytical Instrumentation, Key Laboratory of Bio-resource and Eco-environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, PR China
| | - Yixiang Duan
- Research Center of Analytical Instrumentation, Key Laboratory of Bio-resource and Eco-environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, PR China
| |
Collapse
|
39
|
Wang J, Yu Y, Lu K, Yang M, Li Y, Zhou X, Sun Z. Silica nanoparticles induce autophagy dysfunction via lysosomal impairment and inhibition of autophagosome degradation in hepatocytes. Int J Nanomedicine 2017; 12:809-825. [PMID: 28182147 PMCID: PMC5279829 DOI: 10.2147/ijn.s123596] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Autophagy dysfunction is considered as a potential toxic mechanism of nanomaterials. Silica nanoparticles (SiNPs) can induce autophagy, but the specific mechanism involved remains unclear. Therefore, the aim of this study was to confirm the effects of SiNPs on autophagy dysfunction and explore the possible underlying mechanism. In this article, we reported that cell-internalized SiNPs exhibited dose- and time-dependent cytotoxicity in both L-02 and HepG2 cells. Multiple methods verified that SiNPs induced autophagy even at the noncytotoxic level and blocked the autophagic flux at the high-dose level. Notably, SiNPs impaired the lysosomal function through damaging lysosomal ultrastructures, increasing membrane permeability, and downregulating the expression of lysosomal proteases, cathepsin B, as evidenced by transmission electron microscopy, acridine orange staining, quantitative reverse transcription-polymerase chain reaction, and Western blot assays. Collectively, these data concluded that SiNPs inhibited autophagosome degradation via lysosomal impairment in hepatocytes, resulting in autophagy dysfunction. The current study not only discloses a potential mechanism of autophagy dysfunction induced by SiNPs but also provides novel evidence for the study of toxic effect and safety evaluation of SiNPs.
Collapse
Affiliation(s)
- Ji Wang
- Department of Toxicology and Sanitary Chemistry, School of Public Health; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, People's Republic of China
| | - Yongbo Yu
- Department of Toxicology and Sanitary Chemistry, School of Public Health; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, People's Republic of China
| | - Ke Lu
- Department of Toxicology and Sanitary Chemistry, School of Public Health
| | - Man Yang
- Department of Toxicology and Sanitary Chemistry, School of Public Health; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, People's Republic of China
| | - Yang Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, People's Republic of China
| | - Xianqing Zhou
- Department of Toxicology and Sanitary Chemistry, School of Public Health; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, People's Republic of China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
40
|
Jiang H, Qin XJ, Li WP, Ma R, Wang T, Li ZQ. LncRNAs expression in adjuvant-induced arthritis rats reveals the potential role of LncRNAs contributing to rheumatoid arthritis pathogenesis. Gene 2016; 593:131-142. [PMID: 27511374 DOI: 10.1016/j.gene.2016.08.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 07/28/2016] [Accepted: 08/05/2016] [Indexed: 01/16/2023]
Abstract
BACKGROUND Long non-coding RNAs (LncRNAs) are an important class of widespread molecules involved in diverse biological functions, which are exceptionally expressed in numerous types of diseases. Currently, limited study on LncRNA in rheumatoid arthritis (RA) is available. In this study, we aimed to identify the specifically expressed LncRNA that are relevant to adjuvant-induced arthritis (AA) in rats, and to explore the possible molecular mechanisms of RA pathogenesis. METHODS To identify LncRNAs specifically expressed in rheumatoid arthritis, the expression of LncRNAs in synoviums of rats from the model group (n=3) was compared with that in the control group (n=3) using Arraystar Rat LncRNA/mRNA microarray and real-time polymerase chain reaction (RT-PCR). RESULTS Up to 260 LncRNAs were found to be differentially expressed (≥1.5-fold-change) in the synoviums between AA model and the normal rats (170 up-regulated and 90 down-regulated LncRNAs in AA rats compared with normal rats). Coding-non-coding gene co-expression networks (CNC network) were drawn based on the correlation analysis between the differentially expressed LncRNAs and mRNAs. Six LncRNAs, XR_008357, U75927, MRAK046251, XR_006457, DQ266363 and MRAK003448, were selected to analyze the relationship between LncRNAs and RA via the CNC network and GO analysis. Real-time PCR result confirmed that the six LncRNAs were specifically expressed in the AA rats. CONCLUSIONS These results revealed that clusters of LncRNAs were uniquely expressed in AA rats compared with controls, which manifests that these differentially expressed LncRNAs in AA rats might play a vital role in RA development. Up-regulation or down-regulation of the six LncRNAs might contribute to the molecular mechanism underlying RA. To sum up, our study provides potential targets for treatment of RA and novel profound understanding of the pathogenesis of RA.
Collapse
Affiliation(s)
- Hui Jiang
- College of Basic Medicine, Anhui Medical University, 81 Meishan Road, Hefei, China; Department of Pharmacy, The first affiliated hospital of Anhui university of Chinese medicine, 117 Meishan Road, Hefei, China.
| | - Xiu-Juan Qin
- Department of Pharmacy, The first affiliated hospital of Anhui university of Chinese medicine, 117 Meishan Road, Hefei, China.
| | - Wei-Ping Li
- College of Basic Medicine, Anhui Medical University, 81 Meishan Road, Hefei, China.
| | - Rong Ma
- Department of Integrative Physiology and Cardiovascular Research Institute, University of North Texas Health Sciences Center, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, USA.
| | - Ting Wang
- Department of Pharmacy, The first affiliated hospital of Anhui university of Chinese medicine, 117 Meishan Road, Hefei, China.
| | - Zhu-Qing Li
- College of Basic Medicine, Anhui Medical University, 81 Meishan Road, Hefei, China.
| |
Collapse
|