51
|
Wu N, Li J, Luo H, Wang D, Bai X. Hydroxysafflor yellow A promotes apoptosis via blocking autophagic flux in liver cancer. Biomed Pharmacother 2021; 136:111227. [PMID: 33485070 DOI: 10.1016/j.biopha.2021.111227] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/20/2020] [Accepted: 12/31/2020] [Indexed: 01/27/2023] Open
Abstract
Hydroxysafflor yellow A (HSYA) extracted from the herb Cathartics tinctorius L. negatively regulates liver cancer growth. However, the exact mechanism of HSYA action in liver cancer remains largely unknown. In this study, HSYA inhibited liver cancer cell growth in vivo and in vitro, evidenced by cell proliferation inhibition detected by CCK8, numerous apoptotic cells shown by flow cytometry assay, and expression of apoptosis-related proteins determined by western blot. Importantly, our data revealed that HSYA triggered autophagic response and autophagosome accumulation considering the increased levels of LC3II-conversion examined by western blot, LC3 puncta visualized by immunofluorescence, and expression of autophagy-related genes shown by quantitative real-time PCR. Furthermore, HSYA blocked the late-phase of autophagic flux via impairing the lysosomal acidification and downregulating LAMP1 expression, thereby likely inducing apoptosis. In addition, HSYA inhibited PI3K/AKT/mTOR signaling pathway. Taken together, as HSYA might inhibit cell proliferation and promote apoptosis via blocking autophagic flux in liver cancer, it may be considered a promising candidate for liver cancer therapy.
Collapse
Affiliation(s)
- Na Wu
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, China.
| | - Jingmin Li
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, China.
| | - Hanlin Luo
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, China.
| | - Dong Wang
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, China.
| | - Xianyong Bai
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
52
|
Gao Y, Xu A, Shen Q, Xie Y, Liu S, Wang X. Graphene oxide aggravated dextran sulfate sodium-induced colitis through intestinal epithelial cells autophagy dysfunction. J Toxicol Sci 2021; 46:43-55. [PMID: 33408300 DOI: 10.2131/jts.46.43] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Graphene oxide (GO) is one of the most promising nanomaterials used in biomedicine. However, studies about its adverse effects on the intestine in state of inflammation remain limited. This study aimed to explore the underlying effects of GO on intestinal epithelial cells (IECs) in vitro and colitis in vivo. We found that GO could exert toxic effects on NCM460 cells in a dose- and time-dependent manner and promote inflammation. Furthermore, GO caused lysosomal dysfunction and then blockaded autophagy flux. Moreover, pharmacological autophagy inhibitor 3-Methyladenine could reverse GO-induced LC3B and p62 expression levels, reduce expression levels of IL-6, IL-8, TLR4, and CXCL2, and increase the level of IL-10. In vivo, C57BL/6 mice were treated with 2.5% dextran sulfate sodium (DSS) in drinking water for five consecutive days to induce colitis. Then, GO at 60 mg/kg dose was administered through the oral route every two days from day 2 to day 8. These results showed that GO aggravated DSS-induced colitis, characterized by shortening of the colon and severe pathological changes, and induced autophagy. In conclusion, GO caused the abnormal autophagy in IECs and exacerbated DSS-induced colitis in mice. Our research indicated that GO may contribute to the development of intestinal inflammation by inducing IECs autophagy dysfunction.
Collapse
Affiliation(s)
- Yanfei Gao
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, China
| | - Angao Xu
- Huizhou Medicine Institute, China
| | - Qiong Shen
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, China
| | - Yue Xie
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, China
| | - Siliang Liu
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, China
| | - Xinying Wang
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, China
| |
Collapse
|
53
|
Xiong H, Chen K, Li M. [Role of autophagy in lipopolysaccharide-induced apoptosis of odontoblasts]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:1816-1820. [PMID: 33380391 DOI: 10.12122/j.issn.1673-4254.2020.12.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the role of autophagy in lipopolysaccharide (LPS)-induced apoptosis of murine odontoblasts. METHODS Murine odontoblasts (mDPC-23 cells) were treated with 5 μg/mL LPS for 6, 12 and 24 h, and the changes in cell viability was examined using CCK8 kit and cell apoptosis was detected by TUNEL staining. The changes in the protein levels of LC3, Beclin1, Atg5, AKT, p-AKT, mTOR and p-mTOR were detected using Western blotting. The effect of 3-MA treatment for 24 h on LPS-induced apoptosis of mDPC-23 cells was evaluated by detecting the expressions of apoptosis-related proteins caspase-3 and Bax using Western blotting. RESULTS Stimulation with LPS for 6 and 12 h did not cause significant changes in the proliferation or apoptosis of mDPC-23 cells, but LPS treatment for 24 h significantly suppressed cell proliferation (P < 0.05) and promoted cell apoptosis as shown by TUNEL assay (P < 0.05). Stimulation with LPS for 24 significantly increased the expression levels of LC3, Beclin1 and Atg5, decreased the expressions of p-AKT and p-mTOR (P < 0.05), and obviously upregulated the expressions of caspase-3 and Bax (P < 0.05). Treatment with 3-MA markedly lowered caspase-3 and Bax protein expressions in LPS-stimulated cells (P < 0.05). CONCLUSIONS LPS stimulation induces autophagy to promote apoptosis of mDPC-23 cells, and suppression of autophagy attenuates LPS-induced apoptosis. Autophagy may play an important role in the injury of inflamed pulp tissues.
Collapse
Affiliation(s)
- Huacui Xiong
- Stomatological Hospital, Southern Medical University, Guangzhou 510115, China
| | - Ke Chen
- Stomatological Hospital, Southern Medical University, Guangzhou 510115, China
| | - Meimei Li
- Stomatological Hospital, Southern Medical University, Guangzhou 510115, China
| |
Collapse
|
54
|
Graphene Oxide Ameliorates the Cognitive Impairment Through Inhibiting PI3K/Akt/mTOR Pathway to Induce Autophagy in AD Mouse Model. Neurochem Res 2020; 46:309-325. [PMID: 33180247 DOI: 10.1007/s11064-020-03167-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/31/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease of the central nervous system characterised by cognitive impairment. Its major pathological feature is the deposition of β-amyloid (Aβ) peptide, which triggers a series of pathological cascades. Autophagy is a main pathway to eliminate abnormal aggregated proteins, and increasing autophagy represents a plausible treatment strategy against relative overproduction of neurotoxic Aβ. Graphene oxide (GO) is an emerging carbon-based nanomaterial. As a derivative of graphene with neuroprotective effects, it can effectively increase the clearance of abnormally aggregated protein. In this article, we investigated the protective function of GO in an AD mouse model. GO (30 mg/kg, intraperitoneal) was administered for 2 weeks. The results of the Morris water maze test and the novel object recognition test suggested that GO ameliorated learning and memory impairments in 5xFAD mice. The long-term potentiation and depotentiation from the perforant path to the dentate gyrus in the hippocampus were increased with GO treatment in 5xFAD mice. Furthermore, GO upregulated the expression of synapse-related proteins and increased the cell density in the hippocampus. Our results showed that GO up-regulated LC3II/LC3I and Beclin-1 and decreased p62 protein levels in 5xFAD mice. In addition, GO downregulated the PI3K/Akt/mTOR signalling pathway to induce autophagy. These results have revealed the protective potential of GO in AD.
Collapse
|
55
|
Lim GE, Sung JY, Yu S, Kim Y, Shim J, Kim HJ, Cho ML, Lee JS, Kim YN. Pygenic Acid A (PA) Sensitizes Metastatic Breast Cancer Cells to Anoikis and Inhibits Metastasis In Vivo. Int J Mol Sci 2020; 21:ijms21228444. [PMID: 33182770 PMCID: PMC7696818 DOI: 10.3390/ijms21228444] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/19/2022] Open
Abstract
Metastasis is the main cause of cancer-related deaths. Anoikis is a type of apoptosis caused by cell detachment, and cancer cells become anoikis resistant such that they survive during circulation and can successfully metastasize. Therefore, sensitization of cancer cells to anoikis could prevent metastasis. Here, by screening for anoikis sensitizer using natural compounds, we found that pygenic acid A (PA), a natural compound from Prunella vulgaris, not only induced apoptosis but also sensitized the metastatic triple-negative breast cancer cell lines, MDA-MB-231 cells (human) and 4T1 cells (mouse), to anoikis. Apoptosis protein array and immunoblotting analysis revealed that PA downregulated the pro-survival proteins, including cIAP1, cIAP2, and survivin, leading to cell death of both attached and suspended cells. Interestingly, PA decreased the levels of proteins associated with anoikis resistance, including p21, cyclin D1, p-STAT3, and HO-1. Ectopic expression of active STAT3 attenuated PA-induced anoikis sensitivity. Although PA activated ER stress and autophagy, as determined by increases in the levels of characteristic markers, such as IRE1α, p-elF2α, LC3B I, and LC3B II, PA treatment resulted in p62 accumulation, which could be due to PA-induced defects in autophagy flux. PA also decreased metastatic characteristics, such as cell invasion, migration, wound closure, and 3D growth. Finally, lung metastasis of luciferase-labeled 4T1 cells decreased following PA treatment in a syngeneic mouse model when compared with the control. These data suggest that PA sensitizes metastatic breast cancer cells to anoikis via multiple pathways, such as inhibition of pro-survival pathways and activation of ER stress and autophagy, leading to the inhibition of metastasis. These findings suggest that sensitization to anoikis by PA could be used as a new therapeutic strategy to control the metastasis of breast cancer.
Collapse
Affiliation(s)
- Ga-Eun Lim
- Division of Translational Science, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Korea; (G.-E.L.); (J.Y.S.); (S.Y.); (Y.K.); (J.S.)
| | - Jee Young Sung
- Division of Translational Science, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Korea; (G.-E.L.); (J.Y.S.); (S.Y.); (Y.K.); (J.S.)
| | - Suyeun Yu
- Division of Translational Science, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Korea; (G.-E.L.); (J.Y.S.); (S.Y.); (Y.K.); (J.S.)
| | - Younmi Kim
- Division of Translational Science, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Korea; (G.-E.L.); (J.Y.S.); (S.Y.); (Y.K.); (J.S.)
| | - Jaegal Shim
- Division of Translational Science, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Korea; (G.-E.L.); (J.Y.S.); (S.Y.); (Y.K.); (J.S.)
| | - Hyo Jung Kim
- National Institute for Korean Medicine Development, 94 Hwarang-ro (Gapje-dong), Gyeongsan-si, Gyeongsangbuk-do 38540, Korea; (H.J.K.); (M.L.C.)
| | - Myoung Lae Cho
- National Institute for Korean Medicine Development, 94 Hwarang-ro (Gapje-dong), Gyeongsan-si, Gyeongsangbuk-do 38540, Korea; (H.J.K.); (M.L.C.)
| | - Jae-Seon Lee
- Department of Molecular Medicine, College of Medicine, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Korea;
| | - Yong-Nyun Kim
- Division of Translational Science, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do 10408, Korea; (G.-E.L.); (J.Y.S.); (S.Y.); (Y.K.); (J.S.)
- Correspondence: ; Tel.: +82-31-920-2415; Fax: +82-31-920-2468
| |
Collapse
|
56
|
Qiu B, Bessler N, Figler K, Buchholz M, Rios AC, Malda J, Levato R, Caiazzo M. Bioprinting Neural Systems to Model Central Nervous System Diseases. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1910250. [PMID: 34566552 PMCID: PMC8444304 DOI: 10.1002/adfm.201910250] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/12/2020] [Accepted: 03/16/2020] [Indexed: 05/09/2023]
Abstract
To date, pharmaceutical progresses in central nervous system (CNS) diseases are clearly hampered by the lack of suitable disease models. Indeed, animal models do not faithfully represent human neurodegenerative processes and human in vitro 2D cell culture systems cannot recapitulate the in vivo complexity of neural systems. The search for valuable models of neurodegenerative diseases has recently been revived by the addition of 3D culture that allows to re-create the in vivo microenvironment including the interactions among different neural cell types and the surrounding extracellular matrix (ECM) components. In this review, the new challenges in the field of CNS diseases in vitro 3D modeling are discussed, focusing on the implementation of bioprinting approaches enabling positional control on the generation of the 3D microenvironments. The focus is specifically on the choice of the optimal materials to simulate the ECM brain compartment and the biofabrication technologies needed to shape the cellular components within a microenvironment that significantly represents brain biochemical and biophysical parameters.
Collapse
Affiliation(s)
- Boning Qiu
- Department of PharmaceuticsUtrecht Institute for Pharmaceutical Sciences (UIPS)Utrecht UniversityUniversiteitsweg 99Utrecht3584 CGThe Netherlands
| | - Nils Bessler
- Princess Máxima Center for Pediatric OncologyHeidelberglaan 25Utrecht3584 CSThe Netherlands
| | - Kianti Figler
- Department of PharmaceuticsUtrecht Institute for Pharmaceutical Sciences (UIPS)Utrecht UniversityUniversiteitsweg 99Utrecht3584 CGThe Netherlands
| | - Maj‐Britt Buchholz
- Princess Máxima Center for Pediatric OncologyHeidelberglaan 25Utrecht3584 CSThe Netherlands
| | - Anne C. Rios
- Princess Máxima Center for Pediatric OncologyHeidelberglaan 25Utrecht3584 CSThe Netherlands
| | - Jos Malda
- Department of Orthopaedics and Regenerative Medicine Center UtrechtUniversity Medical Center UtrechtUtrecht UniversityHeidelberglaan 100Utrecht3584CXThe Netherlands
- Department of Equine SciencesFaculty of Veterinary MedicineUtrecht UniversityYalelaan 112Utrecht3584CXThe Netherlands
| | - Riccardo Levato
- Department of Orthopaedics and Regenerative Medicine Center UtrechtUniversity Medical Center UtrechtUtrecht UniversityHeidelberglaan 100Utrecht3584CXThe Netherlands
- Department of Equine SciencesFaculty of Veterinary MedicineUtrecht UniversityYalelaan 112Utrecht3584CXThe Netherlands
| | - Massimiliano Caiazzo
- Department of PharmaceuticsUtrecht Institute for Pharmaceutical Sciences (UIPS)Utrecht UniversityUniversiteitsweg 99Utrecht3584 CGThe Netherlands
- Department of Molecular Medicine and Medical BiotechnologyUniversity of Naples “Federico II”Via Pansini 5Naples80131Italy
| |
Collapse
|
57
|
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
|
58
|
Guo T, Cheng L, Zhao H, Liu Y, Yang Y, Liu J, Wu Q. The C. elegans miR-235 regulates the toxicity of graphene oxide via targeting the nuclear hormone receptor DAF-12 in the intestine. Sci Rep 2020; 10:16933. [PMID: 33037257 PMCID: PMC7547681 DOI: 10.1038/s41598-020-73712-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 09/04/2020] [Indexed: 11/09/2022] Open
Abstract
The increased application of graphene oxide (GO), a new carbon-based engineered nanomaterial, has generated a potential toxicity in humans and the environment. Previous studies have identified some dysregulated microRNAs (miRNAs), such as up-regulated mir-235, in organisms exposed to GO. However, the detailed mechanisms of the dysregulation of miRNA underlying GO toxicity are still largely elusive. In this study, we employed Caenorhabditis elegans as an in vivo model to investigate the biological function and molecular basis of mir-235 in the regulation of GO toxicity. After low concentration GO exposure, mir-235 (n4504) mutant nematodes were sensitive to GO toxicity, implying that mir-235 mediates a protection mechanism against GO toxicity. Tissue-specific assays suggested that mir-235 expressed in intestine is required for suppressing the GO toxicity in C. elegans. daf-12, a gene encoding a member of the steroid hormone receptor superfamily, acts as a target gene of mir-235 in the nematode intestine in response to GO treatment, and RNAi knockdown of daf-12 suppressed the sensitivity of mir-235(n4503) to GO toxicity. Further genetic analysis showed that DAF-12 acted in the upstream of DAF-16 in insulin/IGF-1 signaling pathway and PMK-1 in p38 MAPK signaling pathway in parallel to regulate GO toxicity. Altogether, our results revealed that mir-235 may activate a protective mechanism against GO toxicity by suppressing the DAF-12-DAF-16 and DAF-12-PMK-1 signaling cascade in nematodes, which provides an important molecular basis for the in vivo toxicity of GO at the miRNA level.
Collapse
Affiliation(s)
- Tiantian Guo
- Institute of Nephrology, Zhong Da Hospital, Medical School, Southeast University, Nanjing, China
| | - Lu Cheng
- Institute of Nephrology, Zhong Da Hospital, Medical School, Southeast University, Nanjing, China
| | - Huimin Zhao
- Institute of Nephrology, Zhong Da Hospital, Medical School, Southeast University, Nanjing, China
| | - Yingying Liu
- Institute of Nephrology, Zhong Da Hospital, Medical School, Southeast University, Nanjing, China
| | - Yunhan Yang
- Institute of Nephrology, Zhong Da Hospital, Medical School, Southeast University, Nanjing, China
| | - Jie Liu
- Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Qiuli Wu
- Institute of Nephrology, Zhong Da Hospital, Medical School, Southeast University, Nanjing, China.
| |
Collapse
|
59
|
Liu L, Zhang M, Zhang Q, Jiang W. Graphene nanosheets damage the lysosomal and mitochondrial membranes and induce the apoptosis of RBL-2H3 cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 734:139229. [PMID: 32450398 DOI: 10.1016/j.scitotenv.2020.139229] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/27/2020] [Accepted: 05/03/2020] [Indexed: 06/11/2023]
Abstract
The induced membrane damage is a key mechanism for the cytotoxicity of graphene nanosheets (GNSs). In this research, the physical interaction of GNSs on model membranes was investigated using artificial membranes and plasma membrane vesicles. The effects of the GNSs on plasma membrane, lysosomal and mitochondrial membranes were investigated using rat basophilic leukemia (RBL2H3) cells via lactate dehydrogenase (LDH) assay, acridine orange staining and JC-1 probe, respectively. The physical interaction with model membranes was dominated by electrostatic forces, and the adhered GNSs disrupted the membrane. The degree of physical membrane disruption was quantified by the quartz crystal microbalance with dissipation (QCM-D), confirming the serious membrane disruption. The internalized GNSs were mainly distributed in the lysosomes. They caused plasma membrane leakage, increased the lysosomal membrane permeability (LMP), and depolarized the mitochondrial membrane potential (MMP). The increased cellular levels of reactive oxygen species (ROS) were also detected after GNS exposure. The combination of physical interaction and the excess ROS production damaged the plasma and organelle membranes in living RBL-2H3 cells. The lysosomal and mitochondrial dysfunction, and the oxidative stress further induced cell apoptosis. Specially, the exposure to 25 mg/L GNSs caused severest cell mortality, plasma membrane damage, ROS generation, MMP depolarization and apoptosis. The research findings provide more comprehensive information on the graphene-induced plasma and organelle membrane damage, which is important to understand and predict the cytotoxicity of carbon-based nanomaterials.
Collapse
Affiliation(s)
- Ling Liu
- Environment Research Institute, Shandong University, Qingdao 266237, China
| | - Mengmeng Zhang
- Environment Research Institute, Shandong University, Qingdao 266237, China
| | - Qiu Zhang
- School of Environmental Sciences and Engineering, Shandong University, Qingdao 266237, China
| | - Wei Jiang
- Environment Research Institute, Shandong University, Qingdao 266237, China; Shenzhen Research Institute, Shandong University, Shenzhen 518057, China.
| |
Collapse
|
60
|
Role of autophagy in regulation of cancer cell death/apoptosis during anti-cancer therapy: focus on autophagy flux blockade. Arch Pharm Res 2020; 43:475-488. [PMID: 32458284 DOI: 10.1007/s12272-020-01239-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023]
Abstract
Autophagy is a self-degradation process in which the cytoplasmic cargoes are delivered to the lysosomes for degradation. As the cargoes are degraded/recycled, the autophagy process maintains the cellular homeostasis. Anti-cancer therapies induce apoptosis and autophagy concomitantly, and the induced autophagy normally prevents stress responses that are being induced. In such cases, the inhibition of autophagy can be a reasonable strategy to enhance the efficacy of anti-cancer therapies. However, recent studies have shown that autophagy induced by anti-cancer drugs causes cell death/apoptosis induction, indicating a controversial role of autophagy in cancer cell survival or death/apoptosis. Therefore, in the present review, we aimed to assess the signaling mechanisms involved in autophagy and cell death/apoptosis induction during anti-cancer therapies. This review summarizes the process of autophagy, autophagy flux and its blockade, and measurement and interpretation of autophagy flux. Further, it describes the signaling pathways involved in the blockade of autophagy flux and the role of signaling molecules accumulated by autophagy blockade in cell death/apoptosis in various cancer cells during anti-cancer therapies. Altogether, it implies that factors such as types of cancer, drug therapies, and characteristics of autophagy should be evaluated before targeting autophagy for cancer treatment.
Collapse
|
61
|
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
|
62
|
Abstract
The remarkable advances coming about through nanotechnology promise to revolutionize many aspects of modern life; however, these advances come with a responsibility for due diligence to ensure that they are not accompanied by adverse consequences for human health or the environment. Many novel nanomaterials (having at least one dimension <100 nm) could be highly mobile if released into the environment and are also very reactive, which has raised concerns for potential adverse impacts including, among others, the potential for neurotoxicity. Several lines of evidence led to concerns for neurotoxicity, but perhaps none more than observations that inhaled nanoparticles impinging on the mucosal surface of the nasal epithelium could be internalized into olfactory receptor neurons and transported by axoplasmic transport into the olfactory bulbs without crossing the blood-brain barrier. From the olfactory bulb, there is concern that nanomaterials may be transported deeper into the brain and affect other brain structures. Of course, people will not be exposed to only engineered nanomaterials, but rather such exposures will occur in a complex mixture of environmental materials, some of which are incidentally generated particles of a similar inhalable size range to engineered nanomaterials. To date, most experimental studies of potential neurotoxicity of nanomaterials have not considered the potential exposure sources and pathways that could lead to exposure, and most studies of nanomaterial exposure have not considered potential neurotoxicity. Here, we present a review of potential sources of exposures to nanoparticles, along with a review of the literature on potential neurotoxicity of nanomaterials. We employ the linked concepts of an aggregate exposure pathway (AEP) and an adverse outcome pathway (AOP) to organize and present the material. The AEP includes a sequence of key events progressing from material sources, release to environmental media, external exposure, internal exposure, and distribution to the target site. The AOP begins with toxicant at the target site causing a molecular initiating event and, like the AEP, progress sequentially to actions at the level of the cell, organ, individual, and population. Reports of nanomaterial actions are described at every key event along the AEP and AOP, except for changes in exposed populations that have not yet been observed. At this last stage, however, there is ample evidence of population level effects from exposure to ambient air particles that may act similarly to engineered nanomaterials. The data give an overall impression that current exposure levels may be considerably lower than those reported experimentally to be neurotoxic. This impression, however, is tempered by the absence of long-term exposure studies with realistic routes and levels of exposure to address concerns for chronic accumulation of materials or damage. Further, missing across the board are "key event relationships", which are quantitative expressions linking the key events of either the AEP or the AOP, making it impossible to quantitatively project the likelihood of adverse neurotoxic effects from exposure to nanomaterials or to estimate margins of exposure for such relationships.
Collapse
Affiliation(s)
- William K. Boyes
- Neurological and Endocrine Toxicology Branch, Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC USA 27711
| | - Christoph van Thriel
- Leibniz Research Centre for Working Environment and Human Factors, TU Dortmund, Ardeystr. 67, 44139 Dortmund, Germany
| |
Collapse
|
63
|
Zhao B, Wang H, Dong W, Cheng S, Li H, Tan J, Zhou J, He W, Li L, Zhang J, Luo G, Qian W. A multifunctional platform with single-NIR-laser-triggered photothermal and NO release for synergistic therapy against multidrug-resistant Gram-negative bacteria and their biofilms. J Nanobiotechnology 2020; 18:59. [PMID: 32293461 PMCID: PMC7158002 DOI: 10.1186/s12951-020-00614-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 04/06/2020] [Indexed: 11/10/2022] Open
Abstract
Background Infectious diseases caused by multidrug-resistant (MDR) bacteria, especially MDR Gram-negative strains, have become a global public health challenge. Multifunctional nanomaterials for controlling MDR bacterial infections via eradication of planktonic bacteria and their biofilms are of great interest. Results In this study, we developed a multifunctional platform (TG-NO-B) with single NIR laser-triggered PTT and NO release for synergistic therapy against MDR Gram-negative bacteria and their biofilms. When located at the infected sites, TG-NO-B was able to selectively bind to the surfaces of Gram-negative bacterial cells and their biofilm matrix through covalent coupling between the BA groups of TG-NO-B and the bacterial LPS units, which could greatly improve the antibacterial efficiency, and reduce side damages to ambient normal tissues. Upon single NIR laser irradiation, TG-NO-B could generate hyperthermia and simultaneously release NO, which would synergistically disrupt bacterial cell membrane, further cause leakage and damage of intracellular components, and finally induce bacteria death. On one hand, the combination of NO and PTT could largely improve the antibacterial efficiency. On the other hand, the bacterial cell membrane damage could improve the permeability and sensitivity to heat, decrease the photothermal temperature and avoid damages caused by high temperature. Moreover, TG-NO-B could be effectively utilized for synergistic therapy against the in vivo infections of MDR Gram-negative bacteria and their biofilms and accelerate wound healing as well as exhibit excellent biocompatibility both in vitro and in vivo. Conclusions Our study demonstrates that TG-NO-B can be considered as a promising alternative for treating infections caused by MDR Gram-negative bacteria and their biofilms.
Collapse
Affiliation(s)
- Baohua Zhao
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Key Laboratory of Disease Proteomics of Chongqing, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - He Wang
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Key Laboratory of Disease Proteomics of Chongqing, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Wenjing Dong
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Key Laboratory of Disease Proteomics of Chongqing, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Shaowen Cheng
- Department of Trauma Centre, The First Affiliated Hospital, Hainan Medical University, Haikou, 570102, Hainan, China
| | - Haisheng Li
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Key Laboratory of Disease Proteomics of Chongqing, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jianglin Tan
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Key Laboratory of Disease Proteomics of Chongqing, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Junyi Zhou
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Key Laboratory of Disease Proteomics of Chongqing, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Weifeng He
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Key Laboratory of Disease Proteomics of Chongqing, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Lanlan Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Gaoxing Luo
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Key Laboratory of Disease Proteomics of Chongqing, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Wei Qian
- Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Key Laboratory of Disease Proteomics of Chongqing, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
64
|
Xiaoli F, Qiyue C, Weihong G, Yaqing Z, Chen H, Junrong W, Longquan S. Toxicology data of graphene-family nanomaterials: an update. Arch Toxicol 2020; 94:1915-1939. [DOI: 10.1007/s00204-020-02717-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 03/12/2020] [Indexed: 12/12/2022]
|
65
|
Zhang C, Feng X, He L, Zhang Y, Shao L. The interrupted effect of autophagic flux and lysosomal function induced by graphene oxide in p62-dependent apoptosis of F98 cells. J Nanobiotechnology 2020; 18:52. [PMID: 32188458 PMCID: PMC7081710 DOI: 10.1186/s12951-020-00605-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 03/06/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Graphene oxide (GO) nanoparticles (NPs) have been widely applied in various fields, especially in biomedical applications. Extensive studies have suggested that GO can pass through the blood-brain barrier (BBB) and induce abnormal autophagy and cytotoxicity in the central nervous system (CNS). However, the effect and specific mechanism of GO on astrocytes, the most abundant cells in the brain still has not been extensively investigated. RESULTS In this study, we systematically explored the toxicity and mechanism of GO exposure in the rat astroglioma-derived F98 cell line using molecular biological techniques (immunofluorescence staining, flow cytometry and Western blot) at the subcellular level and the signaling pathway level. Cells exposed to GO exhibited decreased cell viability and increased lactate dehydrogenase (LDH) release in a concentration- and time-dependent manner. GO-induced autophagy was evidenced by transmission electron microscopy (TEM) and immunofluorescence staining. Western blots showed that LC3II/I and p62 were upregulated and PI3K/Akt/mTOR was downregulated. Detection of lysosomal acidity and cathepsin B activity assay indicated the impairment of lysosomal function. Annexin V-FITC-PI detection showed the occurrence of apoptosis after GO exposure. The decrease in mitochondrial membrane potential (MMP) with an accompanying upregulation of cleaved caspase-3 and Bax/Bcl-2 further suggested that endogenous signaling pathways were involved in GO-induced apoptosis. CONCLUSION The exposure of F98 cells to GO can elicit concentration- and time-dependent toxicological effects. Additionally, increased autophagic response can be triggered after GO treatment and that the blocking of autophagy flux plays a vital role in GO cytotoxicity, which was determined to be related to dysfunction of lysosomal degradation. Importantly, the abnormal accumulation of autophagic substrate p62 protein can induce capase-3-mediated apoptosis. Inhibition of abnormal accumulation of autophagic cargo could alleviate the occurrence of GO-induced apoptosis in F98 cells.
Collapse
Affiliation(s)
- Chao Zhang
- Stomatological Hospital, Southern Medical University (Guangdong Provincial Stomatological Hospital), Guangzhou, 510280, China
| | - Xiaoli Feng
- Stomatological Hospital, Southern Medical University (Guangdong Provincial Stomatological Hospital), Guangzhou, 510280, China
| | - Longwen He
- Stomatological Hospital, Southern Medical University (Guangdong Provincial Stomatological Hospital), Guangzhou, 510280, China
| | - Yaqing Zhang
- Stomatological Hospital, Southern Medical University (Guangdong Provincial Stomatological Hospital), Guangzhou, 510280, China
| | - Longquan Shao
- Stomatological Hospital, Southern Medical University (Guangdong Provincial Stomatological Hospital), Guangzhou, 510280, China.
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, China.
| |
Collapse
|