51
|
Franiak-Pietryga I, Ziemba B, Sikorska H, Jander M, Appelhans D, Bryszewska M, Borowiec M. Neurotoxicity of poly(propylene imine) glycodendrimers. Drug Chem Toxicol 2020; 45:1484-1492. [DOI: 10.1080/01480545.2020.1843472] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Ida Franiak-Pietryga
- Department of Clinical and Laboratory Genetics, Medical University of Lodz, Lodz, Poland
- GeneaMed LTD, Lodz, Poland
- University of California San Diego, Moores Cancer Center, San Diego, CA, USA
| | - Barbara Ziemba
- Department of Clinical and Laboratory Genetics, Medical University of Lodz, Lodz, Poland
- GeneaMed LTD, Lodz, Poland
| | | | | | | | - Maria Bryszewska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Maciej Borowiec
- Department of Clinical and Laboratory Genetics, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
52
|
Kulkarni P, Dasgupta P, Bhat NS, Hashimoto Y, Saini S, Shahryari V, Yamamura S, Shiina M, Tanaka Y, Dahiya R, Majid S. Role of the PI3K/Akt pathway in cadmium induced malignant transformation of normal prostate epithelial cells. Toxicol Appl Pharmacol 2020; 409:115308. [PMID: 33129824 DOI: 10.1016/j.taap.2020.115308] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/21/2020] [Accepted: 10/24/2020] [Indexed: 12/28/2022]
Abstract
This study investigated the role of the PI3K/Akt pathway in cadmium (Cd) induced malignant transformation of normal prostate epithelial (PWR1E and RWPE1) cells. Both PWR1E and RWPE1 cells were exposed to 10 μM Cd for one year and designated as Cd-PWR1E and Cd-RWPE1. Cd-RWPE1 cells robustly formed tumors in athymic nude mice. Functionally, Cd-exposure induced tumorigenic attributes indicated by increased wound healing, migration and invasion capabilities in both cell lines. RT2-array analysis revealed many oncogenes including P110α, Akt, mTOR, NFKB1 and RAF were induced whereas tumor suppressor (TS) genes were attenuated in Cd-RWPE1. This was validated by individual quantitative-real-time-PCR at transcriptional and by immunoblot at translational levels. These results were consistent in Cd-PWR1E vs parental PWR1E cells. Gene Set Enrichment Analysis revealed that five prostate cancer (PCa) related pathways were enriched in Cd-exposed cells compared to their normal controls. These pathways include the KEGG- Pathways in cancer, Prostate Cancer Pathway, ERBB, Apoptosis and MAPK pathways. We selected up- and down-regulated genes randomly from the PI3K/Akt pathway array and profiled these in the TCGA/GDC prostate-adenocarcinoma (PRAD) patient cohort. An upregulation of oncogenes and downregulation of TS genes was observed in PCa compared to their normal controls. Taken together, our study reveals that the PI3K/Akt signaling is one of the main molecular pathways involved in Cd-driven transformation of normal prostate epithelial cells to malignant form. Understanding the molecular mechanisms involved in the Cd-driven malignant transformation of normal prostate cells will provide a significant insight to develop better therapeutic strategies for Cd-induced prostate cancer.
Collapse
Affiliation(s)
- Priyanka Kulkarni
- Department of Urology, VA Medical Center and UCSF, San Francisco, CA, USA
| | - Pritha Dasgupta
- Department of Urology, VA Medical Center and UCSF, San Francisco, CA, USA
| | - Nadeem S Bhat
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Yutaka Hashimoto
- Department of Urology, VA Medical Center and UCSF, San Francisco, CA, USA
| | - Sharanjot Saini
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, USA
| | - Varahram Shahryari
- Department of Urology, VA Medical Center and UCSF, San Francisco, CA, USA
| | - Soichiro Yamamura
- Department of Urology, VA Medical Center and UCSF, San Francisco, CA, USA
| | - Marisa Shiina
- Department of Urology, VA Medical Center and UCSF, San Francisco, CA, USA
| | - Yuichiro Tanaka
- Department of Urology, VA Medical Center and UCSF, San Francisco, CA, USA
| | - Rajvir Dahiya
- Department of Urology, VA Medical Center and UCSF, San Francisco, CA, USA.
| | - Shahana Majid
- Department of Urology, VA Medical Center and UCSF, San Francisco, CA, USA.
| |
Collapse
|
53
|
Cholich LA, Pistán ME, Torres AM, Ortega HH, Gardner DR, Bustillo S. Cytotoxic activity induced by the alkaloid extract from Ipomoea carnea on primary murine mixed glial cultures. Toxicon 2020; 188:134-141. [PMID: 33091389 DOI: 10.1016/j.toxicon.2020.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/13/2020] [Accepted: 10/19/2020] [Indexed: 11/15/2022]
Abstract
The prolonged consumption of Ipomoea carnea produces neurologic symptoms in animals and a typical histological lesion, cytoplasmic vacuolization, especially in neurons. The toxic principles of I. carnea are the alkaloids swainsonine and calystegines B1, B2, B3 and C1. In this study, primary brain cultures from newborn mouse containing mixed glial cells were utilized. These cells were exposed to Ipomoea extracts containing between 0 and 250 μM swainsonine for 48 h. Morphological changes were investigated through Phase Contrast microscopy and Rosenfeld's staining. The extract induced cytoplasmic vacuolization in astrocytes and microglia in a dose dependent manner, being more evident when cultures were exposed to 250 μM of swainsonine. In addition, acridine orange staining evidenced an increase in the number of lysosomes in both microglia and astrocytes cells. Consistent with this, scanning electron microscopy also showed that both types of cells presented morphological characteristics of cell activation. Ultrastructurally, cells showed vacuoles filled with amorphous material and surrounded by a single membrane and also multilayer membranes. Taken together, these findings suggest that swainsonine along with calystegines, are probably responsible for the activation of glial cells due to a possible lysosomal dysfunction and therefore intracellular storage. Our results demonstrate that this in vitro glial cell model is a very good alternative to in vivo studies that require several weeks of animal intoxication to observe similar neurotoxic effects.
Collapse
Affiliation(s)
- Luciana Andrea Cholich
- Faculty of Veterinary Science, National University of the Northeast, Corrientes, Argentina; The National Scientific and Technical Research Council (CONICET), Argentina.
| | - María Elena Pistán
- Faculty of Veterinary Science, National University of the Northeast, Corrientes, Argentina; The National Scientific and Technical Research Council (CONICET), Argentina
| | - Ana María Torres
- Natural Products Laboratory, IQUIBA-NEA CONICET, National University of the Northeast, Corrientes, Argentina
| | - Hugo Héctor Ortega
- Institute of Veterinary Sciences of Litoral (ICIVET), National University of Litoral (UNL), Esperanza, Santa Fe, Argentina; The National Scientific and Technical Research Council (CONICET), Argentina
| | - Dale R Gardner
- USDA-ARS Poisonous Plant Research Laboratory, Logan, UT, USA
| | - Soledad Bustillo
- Biological and Molecular Investigations Group (GIBYM), IQUIBA-NEA CONICET, National University of the Northeast, Corrientes, Argentina
| |
Collapse
|
54
|
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
|
55
|
Cui Y, Zhou J, Rong F. Combination of metformin and RG7388 enhances inhibition of growth and induction of apoptosis of ovarian cancer cells through the PI3K/AKT/mTOR pathway. Biochem Biophys Res Commun 2020; 533:665-671. [PMID: 33051060 DOI: 10.1016/j.bbrc.2020.09.135] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 09/29/2020] [Indexed: 10/23/2022]
Abstract
Ovarian cancer is a gynecological cancer that has the highest mortality rate and is often resistant to conventional treatments. Therefore, development of new therapies is essential. Metformin (MET), which is the priority drug for treatment of type 2 diabetes, has received increasing attention because of its anti-tumor effects. Here, we examined combined anti-tumor effects of MET and RG7388, the only MDM2 (mouse double minute 2 homolog) antagonist that has entered phase III clinical trials, on ovarian cancer cell lines. We examined effects on proliferation by Cell Counting Kit-8 (CCK-8) and colony formation assays, and effects on apoptosis by flow cytometric analysis and Hoechst staining. Western blotting was used to measure protein expression in cells and tissues treated with MET and/or RG7388. Flow cytometry was used to measure reactive oxygen species (ROS). We also examined the effects of MET and/or RG7388 on inhibition of A2780 cell growth in vivo. The combination of MET and RG7388 significantly increased growth inhibition, apoptosis, and ROS of A2780 and SKOV3 cells compared with either agent alone. Additionally, in vitro and in vivo results showed that MET and/or RG7388 inhibited the phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway and their combination had a stronger effect. Our findings suggest that the combination of MET and RG7388 enhances growth inhibition and apoptosis induction of ovarian cancer cells through the PI3K/AKT/mTOR pathway and accumulation of intracellular ROS.
Collapse
Affiliation(s)
- Yingying Cui
- Department of Obstetrics and Gynecology, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250014, China
| | - Jing Zhou
- Department of Obstetrics and Gynecology, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250014, China
| | - Fengnian Rong
- Department of Obstetrics and Gynecology, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250014, China.
| |
Collapse
|
56
|
Rahaman MS, Yamasaki S, Binte Hossain KF, Hosokawa T, Saito T, Kurasaki M. Effects of curcumin, D-pinitol alone or in combination in cytotoxicity induced by arsenic in PC12 cells. Food Chem Toxicol 2020; 144:111577. [DOI: 10.1016/j.fct.2020.111577] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/12/2020] [Accepted: 07/01/2020] [Indexed: 12/16/2022]
|
57
|
Malko P, Jiang LH. TRPM2 channel-mediated cell death: An important mechanism linking oxidative stress-inducing pathological factors to associated pathological conditions. Redox Biol 2020; 37:101755. [PMID: 33130440 PMCID: PMC7600390 DOI: 10.1016/j.redox.2020.101755] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/17/2020] [Accepted: 10/08/2020] [Indexed: 12/26/2022] Open
Abstract
Oxidative stress resulting from the accumulation of high levels of reactive oxygen species is a salient feature of, and a well-recognised pathological factor for, diverse pathologies. One common mechanism for oxidative stress damage is via the disruption of intracellular ion homeostasis to induce cell death. TRPM2 is a non-selective Ca2+-permeable cation channel with a wide distribution throughout the body and is highly sensitive to activation by oxidative stress. Recent studies have collected abundant evidence to show its important role in mediating cell death induced by miscellaneous oxidative stress-inducing pathological factors, both endogenous and exogenous, including ischemia/reperfusion and the neurotoxicants amyloid-β peptides and MPTP/MPP+ that cause neuronal demise in the brain, myocardial ischemia/reperfusion, proinflammatory mediators that disrupt endothelial function, diabetogenic agent streptozotocin and diabetes risk factor free fatty acids that induce loss of pancreatic β-cells, bile acids that damage pancreatic acinar cells, renal ischemia/reperfusion and albuminuria that are detrimental to kidney cells, acetaminophen that triggers hepatocyte death, and nanoparticles that injure pericytes. Studies have also shed light on the signalling mechanisms by which these pathological factors activate the TRPM2 channel to alter intracellular ion homeostasis leading to aberrant initiation of various cell death pathways. TRPM2-mediated cell death thus emerges as an important mechanism in the pathogenesis of conditions including ischemic stroke, neurodegenerative diseases, cardiovascular diseases, diabetes, pancreatitis, chronic kidney disease, liver damage and neurovascular injury. These findings raise the exciting perspective of targeting the TRPM2 channel as a novel therapeutic strategy to treat such oxidative stress-associated diseases.
Collapse
Affiliation(s)
- Philippa Malko
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, UK
| | - Lin-Hua Jiang
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province and Department of Physiology and Pathophysiology, Xinxiang Medical University, PR China; School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, UK.
| |
Collapse
|
58
|
Rahaman MS, Banik S, Akter M, Rahman MM, Sikder MT, Hosokawa T, Saito T, Kurasaki M. Curcumin alleviates arsenic-induced toxicity in PC12 cells via modulating autophagy/apoptosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 200:110756. [PMID: 32464442 DOI: 10.1016/j.ecoenv.2020.110756] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/08/2020] [Accepted: 05/09/2020] [Indexed: 06/11/2023]
Abstract
Arsenic is a recognized highly toxic contaminant, responsible for numerous human diseases and affecting many millions of people in different parts of the world. Contrarily, curcumin is a natural dietary polyphenolic compound and the main active ingredient in turmeric. Recently it has drawn great attention due to its diverse biological activities, strong antioxidant properties and therapeutic potential against many human ailments. In this study, we aimed to explore the protective effects and the regulatory role of curcumin on arsenic-induced toxicity and gain insights into biomolecular mechanism/s. Arsenic (10 μM) treatment in PC12 cells for 24 h induced cytotoxicity by decreasing cell viability and intracellular glutathione level and increasing lactate dehydrogenase activity and DNA fragmentation. In addition, arsenic caused apoptotic cell death in PC12 cells, which were confirmed from flow cytometry results. Moreover, arsenic (10 μM) treatment significantly down-regulated the inhibition factors of autophagy/apoptosis; mTOR, Akt, Nrf2, ERK1, Bcl-x, Xiap protein expressions, up-regulated the enhanced factors of autophagy/apoptosis; ULK, LC3, p53, Bax, cytochrome c, caspase 9, cleaved caspase 3 proteins and eventually caused autophagic and apoptotic cell death. However, curcumin (2.5 μM) pretreatment with arsenic (10 μM) effectively saves PC12 cells against arsenic-induced cytotoxicity through increasing cell viability, intracellular GSH level and boosting the antioxidant defense system, and limiting the LDH activity and DNA damage. Furthermore, pretreatment of curcumin with arsenic expressively alleviated arsenic-induced toxicity and cell death by reversing the expressions of proteins; mTOR, Akt, Nrf2, ERK1, Bcl-x, Xiap, ULK, LC3, p53, Bax, cytochrome c, caspase 9 and cleaved caspase 3. Our findings indicated that curcumin showed antioxidant properties through the Nrf2 antioxidant signaling pathway and alleviates arsenic-triggered toxicity in PC12 cells by regulating autophagy/apoptosis.
Collapse
Affiliation(s)
- Md Shiblur Rahaman
- Graduate School of Environmental Science, Hokkaido University, Sapporo, 060-0810, Japan
| | - Subrata Banik
- Graduate School of Environmental Science, Hokkaido University, Sapporo, 060-0810, Japan
| | - Mahmuda Akter
- Graduate School of Environmental Science, Hokkaido University, Sapporo, 060-0810, Japan
| | - Md Mostafizur Rahman
- Department of Environmental Sciences, Jahangirnagar University, Dhaka, 1342, Bangladesh
| | - Md Tajuddin Sikder
- Department of Public Health and Informatics, Jahangirnagar University, Dhaka, 1342, Bangladesh
| | - Toshiyuki Hosokawa
- Institute for the Advancement of Higher Education, Hokkaido University, Sapporo, 060-0817, Japan
| | - Takeshi Saito
- Faculty of Health Sciences, Hokkaido University, Sapporo, 060-0808, Japan
| | - Masaaki Kurasaki
- Graduate School of Environmental Science, Hokkaido University, Sapporo, 060-0810, Japan; Faculty of Environmental Earth Science, Hokkaido University, Sapporo, 060-0810, Japan.
| |
Collapse
|
59
|
Liao C, Jin Y, Li Y, Tjong SC. Interactions of Zinc Oxide Nanostructures with Mammalian Cells: Cytotoxicity and Photocatalytic Toxicity. Int J Mol Sci 2020; 21:E6305. [PMID: 32878253 PMCID: PMC7504403 DOI: 10.3390/ijms21176305] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022] Open
Abstract
This article presents a state-of-the-art review and analysis of literature studies on the morphological structure, fabrication, cytotoxicity, and photocatalytic toxicity of zinc oxide nanostructures (nZnO) of mammalian cells. nZnO with different morphologies, e.g., quantum dots, nanoparticles, nanorods, and nanotetrapods are toxic to a wide variety of mammalian cell lines due to in vitro cell-material interactions. Several mechanisms responsible for in vitro cytotoxicity have been proposed. These include the penetration of nZnO into the cytoplasm, generating reactive oxygen species (ROS) that degrade mitochondrial function, induce endoplasmic reticulum stress, and damage deoxyribonucleic acid (DNA), lipid, and protein molecules. Otherwise, nZnO dissolve extracellularly into zinc ions and the subsequent diffusion of ions into the cytoplasm can create ROS. Furthermore, internalization of nZnO and localization in acidic lysosomes result in their dissolution into zinc ions, producing ROS too in cytoplasm. These ROS-mediated responses induce caspase-dependent apoptosis via the activation of B-cell lymphoma 2 (Bcl2), Bcl2-associated X protein (Bax), CCAAT/enhancer-binding protein homologous protein (chop), and phosphoprotein p53 gene expressions. In vivo studies on a mouse model reveal the adverse impacts of nZnO on internal organs through different administration routes. The administration of ZnO nanoparticles into mice via intraperitoneal instillation and intravenous injection facilitates their accumulation in target organs, such as the liver, spleen, and lung. ZnO is a semiconductor with a large bandgap showing photocatalytic behavior under ultraviolet (UV) light irradiation. As such, photogenerated electron-hole pairs react with adsorbed oxygen and water molecules to produce ROS. So, the ROS-mediated selective killing for human tumor cells is beneficial for cancer treatment in photodynamic therapy. The photoinduced effects of noble metal doped nZnO for creating ROS under UV and visible light for killing cancer cells are also addressed.
Collapse
Affiliation(s)
- Chengzhu Liao
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; (C.L.); (Y.J.)
| | - Yuming Jin
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China; (C.L.); (Y.J.)
| | - Yuchao Li
- Department of Materials Science and Engineering, Liaocheng University, Liaocheng 252000, China
| | - Sie Chin Tjong
- Department of Physics, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| |
Collapse
|
60
|
Chen L, Yu X, Ding H, Zhao Y, Hu C, Feng J. Comparing the Influence of High Doses of Different Zinc Salts on Oxidative Stress and Energy Depletion in IPEC-J2 Cells. Biol Trace Elem Res 2020; 196:481-493. [PMID: 31732928 DOI: 10.1007/s12011-019-01948-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 10/22/2019] [Indexed: 01/06/2023]
Abstract
The current study aimed to investigate the influence of four supplemental zinc salts (chelated: Zn glycine; non-chelated: Zn sulfate, Zn citrate, Zn gluconate) among different zinc concentrations (30-300 μM) on cell proliferation, oxidative stress, and energy depletion in intestinal porcine jejunum epithelial cells (IPEC-J2). Different zinc salts affected cell viability in a time- and dose-dependent manner, which was mainly dependent on the uptake of intracellular Zn2+. Intracellular Zn2+ of Zn sulfate has taken up almost twice as high as Zn glycine when cells were loaded with 100-200 μM zinc. After loading cells with 300 μM zinc, Zn glycine and Zn sulfate had a similar trend in accumulation of Zn2+. When the intracellular Zn2+ overloads, cells will gradually be damaged and subsequently die bearing biochemical features of necrosis or late apoptosis. Meanwhile, obviously, increased levels of intracellular ROS, mitochondrial ROS, MDA, and NO and decreased levels of GSH were observed. Excessive intracellular Zn2+ significantly decreased mitochondria membrane potential accompanied by an obvious loss of ATP and NAD+ levels. Overall, exposure to high doses of zinc salts caused cell damage, which was mainly dependent on the uptake of Zn2+. Zinc overload induced oxidative stress and energy depletion in IPEC-J2 cells, and the cell damage with non-chelated zinc addition was more serious than Zn glycine.
Collapse
Affiliation(s)
- Lingjun Chen
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Xiaonan Yu
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Haoxuan Ding
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yang Zhao
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Caihong Hu
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Jie Feng
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
61
|
Malaviya P, Shukal D, Vasavada AR. Nanotechnology-based Drug Delivery, Metabolism and Toxicity. Curr Drug Metab 2020; 20:1167-1190. [PMID: 31902350 DOI: 10.2174/1389200221666200103091753] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/02/2019] [Accepted: 11/23/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Nanoparticles (NPs) are being used extensively owing to their increased surface area, targeted delivery and enhanced retention. NPs have the potential to be used in many disease conditions. Despite widespread use, their toxicity and clinical safety still remain a major concern. OBJECTIVE The purpose of this study was to explore the metabolism and toxicological effects of nanotherapeutics. METHODS Comprehensive, time-bound literature search was done covering the period from 2010 till date. The primary focus was on the metabolism of NP including their adsorption, degradation, clearance, and bio-persistence. This review also focuses on updated investigations on NPs with respect to their toxic effects on various in vitro and in vivo experimental models. RESULTS Nanotechnology is a thriving field of biomedical research and an efficient drug delivery system. Further their applications are under investigation for diagnosis of disease and as medical devices. CONCLUSION The toxicity of NPs is a major concern in the application of NPs as therapeutics. Studies addressing metabolism, side-effects and safety of NPs are desirable to gain maximum benefits of nanotherapeutics.
Collapse
Affiliation(s)
- Pooja Malaviya
- Department of Cell and Molecular Biology, Iladevi Cataract and IOL Research Centre, Memnagar, Ahmedabad 380052, India.,Ph.D. Scholars, Manipal Academy of Higher Education, Manipal, India
| | - Dhaval Shukal
- Department of Cell and Molecular Biology, Iladevi Cataract and IOL Research Centre, Memnagar, Ahmedabad 380052, India.,Ph.D. Scholars, Manipal Academy of Higher Education, Manipal, India
| | - Abhay R Vasavada
- Department of Cell and Molecular Biology, Iladevi Cataract and IOL Research Centre, Memnagar, Ahmedabad 380052, India
| |
Collapse
|
62
|
Raj EN, Lin Y, Chen C, Liu K, Chao J. Selective Autophagy Pathway of Nanoparticles and Nanodrugs: Drug Delivery and Pathophysiological Effects. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Emmanuel Naveen Raj
- Institute of Molecular Medicine and Bioengineering National Chiao Tung University Hsinchu 30068 Taiwan
- Department of Biological Science and Technology National Chiao Tung University Hsinchu 30068 Taiwan
| | - Yu‐Wei Lin
- Department of Biological Science and Technology National Chiao Tung University Hsinchu 30068 Taiwan
| | - Chien‐Hung Chen
- Department of Biological Science and Technology National Chiao Tung University Hsinchu 30068 Taiwan
| | - Kuang‐Kai Liu
- Department of Biological Science and Technology National Chiao Tung University Hsinchu 30068 Taiwan
| | - Jui‐I Chao
- Institute of Molecular Medicine and Bioengineering National Chiao Tung University Hsinchu 30068 Taiwan
- Department of Biological Science and Technology National Chiao Tung University Hsinchu 30068 Taiwan
- Center For Intelligent Drug Systems and Smart Bio‐devices National Chiao Tung University Hsinchu 30068 Taiwan
| |
Collapse
|
63
|
Jia L, Hao SL, Yang WX. Nanoparticles induce autophagy via mTOR pathway inhibition and reactive oxygen species generation. Nanomedicine (Lond) 2020; 15:1419-1435. [PMID: 32529946 DOI: 10.2217/nnm-2019-0387] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Due to their unique physicochemical properties, nanoparticles (NPs) have been increasingly developed for use in various fields. However, there has been both growing negative concerns with toxicity and positive realization of opportunities in nanomedicine, coming from the growing understanding of the associations between NPs and the human body, particularly relating to their cellular autophagic effects. This review summarizes NP-induced autophagy via the modulation of the mTOR signaling pathway and other associated signals including AMPK and ERK and also demonstrates how reactive oxygen species generation greatly underlies the regulation processes. The perspectives in this review aim to contribute to NP design, particularly in consideration of nanotoxicity and the potential for the precise application of NPs in nanomedicine.
Collapse
Affiliation(s)
- Lu Jia
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Shuang-Li Hao
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, PR China
| |
Collapse
|
64
|
Cho YL, Tan HWS, Saquib Q, Ren Y, Ahmad J, Wahab R, He W, Bay BH, Shen HM. Dual role of oxidative stress-JNK activation in autophagy and apoptosis induced by nickel oxide nanoparticles in human cancer cells. Free Radic Biol Med 2020; 153:173-186. [PMID: 32353482 DOI: 10.1016/j.freeradbiomed.2020.03.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 03/25/2020] [Indexed: 02/08/2023]
Abstract
Nickel oxide nanoparticles (NiO-NPs) are an important group of nanoparticles with increasing applications in many aspects of industry. At present, there is evidence demonstrating the cytotoxic characteristics of NiO-NPs, while the involvement of autophagy in the cytotoxicity of NiO-NPs has not been reported. In this study, we aimed to study the role of autophagy in the cytotoxicity of NiO-NPs and the underlying regulatory mechanisms. First, we provided evidence that NiO-NPs induce autophagy in human cancer cells. Second, we found that the enhanced autophagic flux by NiO-NPs via the generation of intracellular reactive oxygen species (ROS) from mitochondria and the subsequent activation of the JNK pathway. Third, we demonstrated that the activation of JNK is a main force in mediating NiO-NPs-induced apoptosis. Finally, we demonstrated that the autophagic response plays an important protective role against the cytotoxic effect of NiO-NPs. Therefore, this study identifies the dual role of oxidative stress-JNK activation in the biological effects of NiO-NPs via promoting autophagy and mediating apoptosis. Understanding the protective role of autophagy and the underlying mechanism is important for the potential application of NiO-NPs in the biomedical industry.
Collapse
Affiliation(s)
- Yik-Lam Cho
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Hayden Weng Siong Tan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore.
| | - Quaiser Saquib
- Zoology Department, College of Sciences, King Saud University, Riyadh, Saudi Arabia.
| | - Yi Ren
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Javed Ahmad
- Zoology Department, College of Sciences, King Saud University, Riyadh, Saudi Arabia.
| | - Rizwan Wahab
- Zoology Department, College of Sciences, King Saud University, Riyadh, Saudi Arabia.
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Army Medical University, Chongqing, China.
| | - Boon-Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore; Faculty of Health Sciences, University of Macau, Macau.
| |
Collapse
|
65
|
Liu Z, Du Z, Li K, Han Y, Ren G, Yang Z. TRPC6-Mediated Ca 2+ Entry Essential for the Regulation of Nano-ZnO Induced Autophagy in SH-SY5Y Cells. Neurochem Res 2020; 45:1602-1613. [PMID: 32274628 DOI: 10.1007/s11064-020-03025-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 04/02/2020] [Accepted: 04/04/2020] [Indexed: 02/06/2023]
Abstract
Recently, possible applications of zinc oxide nanoparticles (nano-ZnO) have been extensively studied owing to their ease of synthesis. However, the effect of nano-ZnO on the nervous system remains unclear. This study investigates the action of nano-ZnO on SH-SY5Y neuroblastoma cells. We found that nano-ZnO (0-50 µg/mL) induced a significant decrease in cell survival rate in a dose-dependent manner, and increased LC3 puncta formation. However, the apoptosis was not affected by nano-ZnO, because the protein levels of cytochrome c, caspase-3, Bcl-xL, and BAX were not varied by the nano-ZnO treatment. Nano-ZnO increased Ca2+ entry and the expression of TRPC6.The results suggested that nano-ZnO increased [Ca2+] through the TRPC-dependent Ca2+ influx, since Ca2+ influx can be prevented by the TRPC inhibitor. Furthermore, cells on nano-ZnO-treatment groups displayed loss of F-actin in a dose dependent manner, which also could be prevented by TRPC inhibitor. Herein, we demonstrated that the nano-ZnO activated TRPC6 channel, thereby increasing the Ca2+ flux and resulting in increased autophagy. Nano-ZnO could have possible anticancer effects in neuroblastoma by inhibiting the proliferation of tumor cells. However, we should also pay attention toward the biosecurity of nano materials.
Collapse
Affiliation(s)
- Zhaowei Liu
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, No.94, Weijin Road, Nankai District, Tianjin, 300071, China.
| | - Zhanqiang Du
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, No.94, Weijin Road, Nankai District, Tianjin, 300071, China
| | - Kai Li
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, No.94, Weijin Road, Nankai District, Tianjin, 300071, China
| | - Yangguang Han
- School of precision instrument and optoelectronic engineering, Tianjin University, Tianjin, 300072, China
| | - Guogang Ren
- Science and Technology Research Institute, University of Hertfordshire, Hatfield, Herts, AL10 9AB, UK
| | - Zhuo Yang
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, No.94, Weijin Road, Nankai District, Tianjin, 300071, China.
| |
Collapse
|
66
|
Wang L, Yin YL, Liu XZ, Shen P, Zheng YG, Lan XR, Lu CB, Wang JZ. Current understanding of metal ions in the pathogenesis of Alzheimer's disease. Transl Neurodegener 2020; 9:10. [PMID: 32266063 PMCID: PMC7119290 DOI: 10.1186/s40035-020-00189-z] [Citation(s) in RCA: 257] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 03/11/2020] [Indexed: 02/07/2023] Open
Abstract
Background The homeostasis of metal ions, such as iron, copper, zinc and calcium, in the brain is crucial for maintaining normal physiological functions. Studies have shown that imbalance of these metal ions in the brain is closely related to the onset and progression of Alzheimer's disease (AD), the most common neurodegenerative disorder in the elderly. Main body Erroneous deposition/distribution of the metal ions in different brain regions induces oxidative stress. The metal ions imbalance and oxidative stress together or independently promote amyloid-β (Aβ) overproduction by activating β- or γ-secretases and inhibiting α-secretase, it also causes tau hyperphosphorylation by activating protein kinases, such as glycogen synthase kinase-3β (GSK-3β), cyclin-dependent protein kinase-5 (CDK5), mitogen-activated protein kinases (MAPKs), etc., and inhibiting protein phosphatase 2A (PP2A). The metal ions imbalances can also directly or indirectly disrupt organelles, causing endoplasmic reticulum (ER) stress; mitochondrial and autophagic dysfunctions, which can cause or aggravate Aβ and tau aggregation/accumulation, and impair synaptic functions. Even worse, the metal ions imbalance-induced alterations can reversely exacerbate metal ions misdistribution and deposition. The vicious cycles between metal ions imbalances and Aβ/tau abnormalities will eventually lead to a chronic neurodegeneration and cognitive deficits, such as seen in AD patients. Conclusion The metal ions imbalance induces Aβ and tau pathologies by directly or indirectly affecting multiple cellular/subcellular pathways, and the disrupted homeostasis can reversely aggravate the abnormalities of metal ions transportation/deposition. Therefore, adjusting metal balance by supplementing or chelating the metal ions may be potential in ameliorating AD pathologies, which provides new research directions for AD treatment.
Collapse
Affiliation(s)
- Lu Wang
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Ya-Ling Yin
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Xin-Zi Liu
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Peng Shen
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Yan-Ge Zheng
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Xin-Rui Lan
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Cheng-Biao Lu
- 1Key Laboratory of Brain Research of Henan Province, Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, 453003 China
| | - Jian-Zhi Wang
- 2Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| |
Collapse
|
67
|
Hernández-Camacho JD, Vicente-García C, Parsons DS, Navas-Enamorado I. Zinc at the crossroads of exercise and proteostasis. Redox Biol 2020; 35:101529. [PMID: 32273258 PMCID: PMC7284914 DOI: 10.1016/j.redox.2020.101529] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 12/11/2022] Open
Abstract
Zinc is an essential element for all forms of life, and one in every ten human proteins is a zinc protein. Zinc has catalytic, structural and signalling functions and its correct homeostasis affects many cellular processes. Zinc deficiency leads to detrimental consequences, especially in tissues with high demand such as skeletal muscle. Zinc cellular homeostasis is tightly regulated by different transport and buffer protein systems. Specifically, in skeletal muscle, zinc has been found to affect myogenesis and muscle regeneration due to its effects on muscle cell activation, proliferation and differentiation. In relation to skeletal muscle, exercise has been shown to modulate zinc serum and urinary levels and could directly affect cellular zinc transport. The oxidative stress induced by exercise may provide the basis for the mild zinc deficiency observed in athletes and could have severe consequences on health and sport performance. Proteostasis is induced during exercise and zinc plays an essential role in several of the associated pathways. Zinc deficiency could be a crucial issue in sport performance for athletes. Exercise could modulate zinc serum and cellular homeostasis. Zinc is part of proteostatic systems critical during exercise.
Collapse
Affiliation(s)
- Juan Diego Hernández-Camacho
- Centro Andaluz de Biología del Desarrollo, CSIC-UPO-JA, Universidad Pablo de Olavide, Sevilla, 41013, Spain; CIBERER, Instituto de Salud Carlos III, Madrid, 28000, Spain
| | - Cristina Vicente-García
- Centro Andaluz de Biología del Desarrollo, CSIC-UPO-JA, Universidad Pablo de Olavide, Sevilla, 41013, Spain
| | | | | |
Collapse
|
68
|
Navarro-Palomares E, González-Saiz P, Renero-Lecuna C, Martín-Rodríguez R, Aguado F, González-Alonso D, Fernández Barquín L, González J, Bañobre-López M, Fanarraga ML, Valiente R. Dye-doped biodegradable nanoparticle SiO 2 coating on zinc- and iron-oxide nanoparticles to improve biocompatibility and for in vivo imaging studies. NANOSCALE 2020; 12:6164-6175. [PMID: 32133463 DOI: 10.1039/c9nr08743e] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
In vivo imaging and therapy represent one of the most promising areas in nanomedicine. Particularly, the identification and localization of nanomaterials within cells and tissues are key issues to understand their interaction with biological components, namely their cell internalization route, intracellular destination, therapeutic activity and possible cytotoxicity. Here, we show the development of multifunctional nanoparticles (NPs) by providing luminescent functionality to zinc and iron oxide NPs. We describe simple synthesis methods based on modified Stöber procedures to incorporate fluorescent molecules on the surface of oxide NPs. These procedures involve the successful coating of NPs with size-controlled amorphous silica (SiO2) shells incorporating standard chromophores like fluorescein, rhodamine B or rhodamine B isothiocyanate. Specifically, spherical Fe3O4 NPs with an average size of 10 nm and commercial ZnO NPs (ca. 130 nm), both coated with an amorphous SiO2 shell of ca. 15 and 24 nm thickness, respectively, are presented. The magnetic nanoparticles, with a major presence of magnetite, show negligible coercitivity. Hence, interactions (dipolar) are very weak and the cores are in the superparamagnetic regime. Spectroscopic measurements confirm the presence of fluorescent molecules within the SiO2 shell, making these hybrid NPs suitable for bioimaging. Thus, our coating procedures improve NP dispersibility in physiological media and allow the identification and localization of intracellular ZnO and Fe3O4 NPs using confocal microscopy imaging preserving the fluorescence of the NP. We demonstrate how both Fe3O4 and ZnO NPs coated with luminescent SiO2 are internalized and accumulated in the cell cytoplasm after 24 hours. Besides, the SiO2 shell provides a platform for further functionalization that enables the design of targeted therapeutic strategies. Finally, we studied the degradation of the shell in different physiological environments, pointing out that the SiO2 coating is stable enough to reach the target cells maintaining its original structure. Degradation took place only 24 hours after exposure to different media.
Collapse
Affiliation(s)
- Elena Navarro-Palomares
- Dpto. de Física Aplicada, Universidad de Cantabria, Facultad de Ciencias, 39005 Santander, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
"Iron free" zinc oxide nanoparticles with ion-leaking properties disrupt intracellular ROS and iron homeostasis to induce ferroptosis. Cell Death Dis 2020; 11:183. [PMID: 32170066 PMCID: PMC7070056 DOI: 10.1038/s41419-020-2384-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 12/22/2022]
Abstract
Exposure to nanomaterials (NMs) is an emerging threat to human health, and the understanding of their intracellular behavior and related toxic effects is urgently needed. Ferroptosis is a newly discovered, iron-mediated cell death that is distinctive from apoptosis or other cell-death pathways. No evidence currently exists for the effect of “iron free” engineered NMs on ferroptosis. We showed by several approaches that (1) zinc oxide nanoparticles (ZnO NPs)-induced cell death involves ferroptosis; (2) ZnO NPs-triggered ferroptosis is associated with elevation of reactive oxygen species (ROS) and lipid peroxidation, along with depletion of glutathione (GSH) and downregulation of glutathione peroxidase 4 (GPx4); (3) ZnO NPs disrupt intracellular iron homeostasis by orchestrating iron uptake, storage and export; (4) p53 largely participates in ZnO NPs-induced ferroptosis; and (5) ZnO particle remnants and dissolved zinc ion both contribute to ferroptosis. In conclusion, our data provide a new mechanistic rationale for ferroptosis as a novel cell-death phenotype induced by engineered NMs.
Collapse
|
70
|
Nabeel AI. Samarium enriches antitumor activity of ZnO nanoparticles via downregulation of CXCR4 receptor and cytochrome P450. Tumour Biol 2020; 42:1010428320909999. [PMID: 32129155 DOI: 10.1177/1010428320909999] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Cancer is the leading cause of death and exhausts human and economic resources for treatment and protection. Zinc oxide nanoparticles play an effective role in tumor treatment but with some cautions, such as overexpression of cytochrome P450, hepatic overload, and the mammalian target of rapamycin pathway resistance. Although lanthanides have antitumor activity, their use is limited. Therefore, the current study aims to improve the effectiveness of zinc oxide nanoparticle via doping with lanthanides, such as samarium. In vitro study revealed that samarium doped with zinc oxide showed more antitumor activity than the other lanthanides, and the antitumor activity depends on the concentration of samarium in the nanocomposite. The in vivo experiment on mice bearing Ehrlich solid tumor revealed that intramuscular injection of samarium/zinc oxide downregulates the expressions of CXCR4 and PI3K/Akt/mammalian target of rapamycin pathway in respect to Ehrlich solid tumor group. Regarding the apoptotic biomarkers, samarium/zinc oxide upregulates the apoptotic biomarker; Bax accompanied with the mitotic catastrophe which was indicated by cell cycle arrest in G2 phase. Moreover, samarium:zinc oxide nanoparticles exhibited minimum toxicity which was indicated by suppressed activities of cytochrome P450 and hepatic enzymes, including alanine transaminase and aspartate transaminase. In addition, the histopathological finding, as well as immunophenotyping results, appreciated the biochemical finding. Therefore, samarium:zinc oxide might be offered a new approach to improve the effectiveness of zinc oxide nanoparticles along with lower toxic effect. Also, samarium:zinc oxide nanoparticles can be a candidate as a new antitumor compound to detect its mode of action.
Collapse
Affiliation(s)
- Asmaa I Nabeel
- Biochemistry Laboratory, Chemistry Department, Faculty of Education, Ain Shams University, Cairo, Egypt
| |
Collapse
|
71
|
Nanoparticle-Mediated Therapeutic Application for Modulation of Lysosomal Ion Channels and Functions. Pharmaceutics 2020; 12:pharmaceutics12030217. [PMID: 32131531 PMCID: PMC7150957 DOI: 10.3390/pharmaceutics12030217] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 02/07/2023] Open
Abstract
Applications of nanoparticles in various fields have been addressed. Nanomaterials serve as carriers for transporting conventional drugs or proteins through lysosomes to various cellular targets. The basic function of lysosomes is to trigger degradation of proteins and lipids. Understanding of lysosomal functions is essential for enhancing the efficacy of nanoparticles-mediated therapy and reducing the malfunctions of cellular metabolism. The lysosomal function is modulated by the movement of ions through various ion channels. Thus, in this review, we have focused on the recruited ion channels for lysosomal function, to understand the lysosomal modulation through the nanoparticles and its applications. In the future, lysosomal channels-based targets will expand the therapeutic application of nanoparticles-associated drugs.
Collapse
|
72
|
Hirano S, Kanno S. Relevance of autophagy markers to cytotoxicity of zinc compounds in macrophages. Toxicol In Vitro 2020; 65:104816. [PMID: 32126253 DOI: 10.1016/j.tiv.2020.104816] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/28/2020] [Indexed: 10/24/2022]
Abstract
Autophagy molecules such as microtubule-associated protein light chain 3 (LC3) and p62/SQSTM1 have been used as biomarkers of protective or conversely adverse effects of exposure to toxicants. In the present study we show changes in LC3-II (a lipidated form of LC3-I) and p62 levels in response to zinc compounds and some other toxicants in J774.1 murine macrophages. The cytotoxicity of either ZnO or ZnSO4 largely depended on the concentration of FBS or albumin in the culture medium. Accordingly, these authophagy markers were more remarkably increased when the cells were exposed to ZnO or ZnSO4 in the absence of FBS. We next addressed lysosomal function impairment and changes in LC3-II and p62 levels following exposure to TiO2, ZnO, and ZnSO4. Lysosomal pH was quickly decreased by autolysosome inhibitors such as bafilomycin A1 and chloroquine, while TiO2, ZnO and ZnSO4 did not decrease lysosomal pH. However, the amounts of LC3-II and p62 and the LC3-II/LC3-I ratio were increased either by the lysosomal inhibitors and the Zn compounds. LC3-II and p62 levels were increased after exposure to arsenite and lipopolysaccharide (LPS). The p62 and phospho-p62 levels were also increased by either ZnSO4 and bafilomycin A1 in HEK293 cells stably expressing RFP-LC3. The current observations suggest that LC3-II and p62 levels were increased as consequences of early effects of toxicants without changing lysosomal pH.
Collapse
Affiliation(s)
- Seishiro Hirano
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, Ibaraki 305-8506, Japan.
| | - Sanae Kanno
- Department of Forensic Medicine, Nagoya City University Graduate School of Medical Sciences, Japan
| |
Collapse
|
73
|
Wang Y, Zhao H, Guo M, Fei D, Zhang L, Xing M. Targeting the miR-122/PKM2 autophagy axis relieves arsenic stress. JOURNAL OF HAZARDOUS MATERIALS 2020; 383:121217. [PMID: 31546213 DOI: 10.1016/j.jhazmat.2019.121217] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 08/12/2019] [Accepted: 09/11/2019] [Indexed: 06/10/2023]
Abstract
Arsenic (As) is a natural hepatotoxicity inducer that is found ubiquitously in foods and environmental media. We found that arsenite exposure elicits autophagy in vivo and vitro, the specific role and regulatory mechanism of which are yet clear. MicroRNAs (miRNAs) are short noncoding RNAs that function in the posttranscriptional regulation of gene expression. Here, we report that miR-122, the most enriched constitutive miRNA in the liver, induced cell protective autophagy in arsenite-exposed hepatocytes. Arsenite exposure elevated miRNA-122 level and decreased the level of its target gene, PKM2. Under arsenic stress, overexpression of miR-122 significantly induced cell protective autophagy, characterized by lipidation of LC3-II and a corresponding consumption of p62. Conversely, autophagy inhibition by miR-122 knockdown was reversed by si-PKM2 cotransfection. We also found that miR-122 knockdown positively regulated the PI3K/Akt/mTOR pathway, and this phenomenon was reversed by cotransfecting cells with si-PKM2. Taken together, our findings show that the miR-122/PKM2 autophagy axis protects hepatocytes from arsenite stress via the PI3K/Akt/mTOR pathway; thus, miR-122 may be a potential candidate in the treatment of arseniasis.
Collapse
Affiliation(s)
- Yu Wang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China.
| | - Hongjing Zhao
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China.
| | - Menghao Guo
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China
| | - Dongxue Fei
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China
| | - Lina Zhang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China
| | - Mingwei Xing
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang, PR China.
| |
Collapse
|
74
|
Rahaman MS, Akter M, Rahman MM, Sikder MT, Hosokawa T, Saito T, Kurasaki M. Investigating the protective actions of D-pinitol against arsenic-induced toxicity in PC12 cells and the underlying mechanism. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 74:103302. [PMID: 31786496 DOI: 10.1016/j.etap.2019.103302] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 06/10/2023]
Abstract
Arsenic is awfully toxic metalloid responsible for many human diseases all over the world. Contrastingly, D-pinitol is a naturally occurring bioactive dietary compound has antioxidant properties. The objective of this study is to elucidate the protective actions of D-pinitol on arsenic-induced cytotoxicity and explore its controlling role in biomolecular mechanisms in PC12 cells. Obtained results demonstrated that co-exposure of D-pinitol with arsenic increases cell viability, decreases DNA damage and protects PC12 cells from arsenic-induced cytotoxicity by increasing glutathione (GSH) level and glutathione reductase (GR). Protein expression of western blot analysis showed that co-exposure of D-pinitol and arsenic significantly inhibited arsenic-induced autophagy which further suppressed apoptosis through up-regulation of survival factors; mTOR, p-mTOR, Akt, p-Akt, NF-кB, Nrf2, ERK1, GR, Bcl-x and down-regulation of death factors; p53, Bax, cytochrome c, LC3, although arsenic regulated those factors negatively. These results of this study suggested that D-pinitol protects PC12 cells from arsenic-induced cytotoxicity.
Collapse
Affiliation(s)
- Md Shiblur Rahaman
- Graduate School of Environmental Science, Hokkaido University, Sapporo, 060-0810, Japan
| | - Mahmuda Akter
- Graduate School of Environmental Science, Hokkaido University, Sapporo, 060-0810, Japan
| | - Md Mostafizur Rahman
- Department of Environmental Sciences, Jahangirnagar University, Dhaka, 1342, Bangladesh
| | - Md Tajuddin Sikder
- Department of Public Health and Informatics, Jahangirnagar University, Dhaka, 1342, Bangladesh
| | - Toshiyuki Hosokawa
- Institute for the Advancement of Higher Education, Hokkaido University, Sapporo, 060-0817, Japan
| | - Takeshi Saito
- Faculty of Health Sciences, Hokkaido University, Sapporo, 060-0808, Japan
| | - Masaaki Kurasaki
- Graduate School of Environmental Science, Hokkaido University, Sapporo, 060-0810, Japan; Faculty of Environmental Earth Science, Hokkaido University, Sapporo, 060-0810, Japan.
| |
Collapse
|
75
|
Wiesmann N, Tremel W, Brieger J. Zinc oxide nanoparticles for therapeutic purposes in cancer medicine. J Mater Chem B 2020; 8:4973-4989. [DOI: 10.1039/d0tb00739k] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Zinc oxide nanoparticles are characterized by a good biocompatibility while providing a versatile potential as innovative therapeutic agents in cancer medicine.
Collapse
Affiliation(s)
- Nadine Wiesmann
- Department of Otorhinolaryngology, Head and Neck Surgery
- University Medical Centre of the Johannes Gutenberg-University
- Laboratory for Molecular Tumor Biology
- 55131 Mainz
- Germany
| | - Wolfgang Tremel
- Department of Chemistry
- Johannes Gutenberg-University
- 55128 Mainz
- Germany
| | - Juergen Brieger
- Department of Otorhinolaryngology, Head and Neck Surgery
- University Medical Centre of the Johannes Gutenberg-University
- Laboratory for Molecular Tumor Biology
- 55131 Mainz
- Germany
| |
Collapse
|
76
|
Banik S, Akter M, Corpus Bondad SE, Saito T, Hosokawa T, Kurasaki M. Carvacrol inhibits cadmium toxicity through combating against caspase dependent/independent apoptosis in PC12 cells. Food Chem Toxicol 2019; 134:110835. [DOI: 10.1016/j.fct.2019.110835] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 09/19/2019] [Accepted: 09/21/2019] [Indexed: 01/03/2023]
|
77
|
Alghsham RS, Satpathy SR, Bodduluri SR, Hegde B, Jala VR, Twal W, Burlison JA, Sunkara M, Haribabu B. Zinc Oxide Nanowires Exposure Induces a Distinct Inflammatory Response via CCL11-Mediated Eosinophil Recruitment. Front Immunol 2019; 10:2604. [PMID: 31787980 PMCID: PMC6856074 DOI: 10.3389/fimmu.2019.02604] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/21/2019] [Indexed: 01/21/2023] Open
Abstract
High aspect ratio zinc oxide nanowires (ZnONWs) have become one of the most important products in nanotechnology. The wide range applications of ZnONWs have heightened the need for evaluating the risks and biological consequences to these particles. In this study, we investigated inflammatory pathways activated by ZnONWs in cultured cells as well as the consequences of systemic exposure in mouse models. Confocal microscopy showed rapid phagocytic uptake of FITC-ZnONWs by macrophages. Exposure of macrophages or lung epithelial cells to ZnONWs induced the production of CCL2 and CCL11. Moreover, ZnONWs exposure induced both IL-6 and TNF-α production only in macrophages but not in LKR13 cells. Intratracheal instillation of ZnONWs in C57BL/6 mice induced a significant increase in the total numbers of immune cells in the broncho alveolar lavage fluid (BALFs) 2 days after instillation. Macrophages and eosinophils were the predominant cellular infiltrates of ZnONWs exposed mouse lungs. Similar cellular infiltrates were also observed in a mouse air-pouch model. Pro-inflammatory cytokines IL-6 and TNF-α as well as chemokines CCL11, and CCL2 were increased both in BALFs and air-pouch lavage fluids. These results suggest that exposure to ZnONWs may induce distinct inflammatory responses through phagocytic uptake and formation of soluble Zn2+ ions.
Collapse
Affiliation(s)
- Ruqaih S Alghsham
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States.,James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Shuchismita R Satpathy
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States.,James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Sobha R Bodduluri
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States.,James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Bindu Hegde
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States.,James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Venkatakrishna R Jala
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States.,James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Waleed Twal
- Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Joseph A Burlison
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Mahendra Sunkara
- Department of Chemical Engineering, Conn Center for Renewable Energy, University of Louisville, Louisville, KY, United States
| | - Bodduluri Haribabu
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States.,James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| |
Collapse
|
78
|
Li F, Li Z, Jin X, Liu Y, Li P, Shen Z, Wu A, Zheng X, Chen W, Li Q. Radiosensitizing Effect of Gadolinium Oxide Nanocrystals in NSCLC Cells Under Carbon Ion Irradiation. NANOSCALE RESEARCH LETTERS 2019; 14:328. [PMID: 31637533 PMCID: PMC6803611 DOI: 10.1186/s11671-019-3152-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 09/09/2019] [Indexed: 05/17/2023]
Abstract
Gadolinium-based nanomaterials can not only serve as contrast agents but also contribute to sensitization in the radiotherapy of cancers. Among radiotherapies, carbon ion irradiation is considered one of the superior approaches with unique physical and biological advantages. However, only a few metallic nanoparticles have been used to improve carbon ion irradiation. In this study, gadolinium oxide nanocrystals (GONs) were synthesized using a polyol method to decipher the radiosensitizing mechanisms in non-small cell lung cancer (NSCLC) cell lines irradiated by carbon ions. The sensitizer enhancement ratio at the 10% survival level was correlated with the concentration of Gd in NSCLC cells. GONs elicited an increase in hydroxyl radical production in a concentration-dependent manner, and the yield of reactive oxygen species increased obviously in irradiated cells, which led to DNA damage and cell cycle arrest. Apoptosis and cytostatic autophagy were also significantly induced by GONs under carbon ion irradiation. The GONs may serve as an effective theranostic material in carbon ion radiotherapy for NSCLC.
Collapse
Affiliation(s)
- Feifei Li
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000 Gansu Province China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Zihou Li
- University of Chinese Academy of Sciences, Beijing, 100049 China
- Key Laboratory of Magnetic Materials and Devices, Chinese Academy of Sciences, Division of Functional Materials and Nano Devices, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo, 315201 Zhejiang China
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000 Gansu Province China
| | - Yan Liu
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000 Gansu Province China
| | - Ping Li
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000 Gansu Province China
| | - Zheyu Shen
- Key Laboratory of Magnetic Materials and Devices, Chinese Academy of Sciences, Division of Functional Materials and Nano Devices, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo, 315201 Zhejiang China
| | - Aiguo Wu
- Key Laboratory of Magnetic Materials and Devices, Chinese Academy of Sciences, Division of Functional Materials and Nano Devices, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences, Ningbo, 315201 Zhejiang China
| | - Xiaogang Zheng
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000 Gansu Province China
| | - Weiqiang Chen
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000 Gansu Province China
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, 730000 Gansu Province China
| |
Collapse
|
79
|
DeLong RK, Cheng YH, Pearson P, Lin Z, Coffee C, Mathew EN, Hoffman A, Wouda RM, Higginbotham ML. Translating Nanomedicine to Comparative Oncology-the Case for Combining Zinc Oxide Nanomaterials with Nucleic Acid Therapeutic and Protein Delivery for Treating Metastatic Cancer. J Pharmacol Exp Ther 2019; 370:671-681. [PMID: 31040175 PMCID: PMC6806346 DOI: 10.1124/jpet.118.256230] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/04/2019] [Indexed: 01/16/2023] Open
Abstract
The unique anticancer, biochemical, and immunologic properties of nanomaterials are becoming a new tool in biomedical research. Their translation into the clinic promises a new wave of targeted therapies. One nanomaterial of particular interest are zinc oxide (ZnO) nanoparticles (NPs), which has distinct mechanisms of anticancer activity including unique surface, induction of reactive oxygen species, lipid oxidation, pH, and also ionic gradients within cancer cells and the tumor microenvironment. It is recognized that ZnO NPs can serve as a direct enzyme inhibitor. Significantly, ZnO NPs inhibit extracellular signal-regulated kinase (ERK) and protein kinase B (AKT) associated with melanoma progression, drug resistance, and metastasis. Indeed, direct intratumoral injection of ZnO NPs or a complex of ZnO with RNA significantly suppresses ERK and AKT phosphorylation. These data suggest ZnO NPs and their complexes or conjugates with nucleic acid therapeutic or anticancer protein may represent a potential new strategy for the treatment of metastatic melanoma, and potentially other cancers. This review focuses on the anticancer mechanisms of ZnO NPs and what is currently known about its biochemical effects on melanoma, biologic activity, and pharmacokinetics in rodents and its potential for translation into large animal, spontaneously developing models of melanoma and other cancers, which represent models of comparative oncology.
Collapse
Affiliation(s)
- R K DeLong
- Department of Anatomy and Physiology, Nanotechnology Innovation Center (R.K.D., P.P., E.N.M., A.H.), Department of Anatomy and Physiology, Institute for Computational Comparative Medicine (Y.-H.C., Z.L.), and Department of Clinical Sciences (C.C., R.M.W., M.L.H.), College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Yi-Hsien Cheng
- Department of Anatomy and Physiology, Nanotechnology Innovation Center (R.K.D., P.P., E.N.M., A.H.), Department of Anatomy and Physiology, Institute for Computational Comparative Medicine (Y.-H.C., Z.L.), and Department of Clinical Sciences (C.C., R.M.W., M.L.H.), College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Paige Pearson
- Department of Anatomy and Physiology, Nanotechnology Innovation Center (R.K.D., P.P., E.N.M., A.H.), Department of Anatomy and Physiology, Institute for Computational Comparative Medicine (Y.-H.C., Z.L.), and Department of Clinical Sciences (C.C., R.M.W., M.L.H.), College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Zhoumeng Lin
- Department of Anatomy and Physiology, Nanotechnology Innovation Center (R.K.D., P.P., E.N.M., A.H.), Department of Anatomy and Physiology, Institute for Computational Comparative Medicine (Y.-H.C., Z.L.), and Department of Clinical Sciences (C.C., R.M.W., M.L.H.), College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Calli Coffee
- Department of Anatomy and Physiology, Nanotechnology Innovation Center (R.K.D., P.P., E.N.M., A.H.), Department of Anatomy and Physiology, Institute for Computational Comparative Medicine (Y.-H.C., Z.L.), and Department of Clinical Sciences (C.C., R.M.W., M.L.H.), College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Elza Neelima Mathew
- Department of Anatomy and Physiology, Nanotechnology Innovation Center (R.K.D., P.P., E.N.M., A.H.), Department of Anatomy and Physiology, Institute for Computational Comparative Medicine (Y.-H.C., Z.L.), and Department of Clinical Sciences (C.C., R.M.W., M.L.H.), College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Amanda Hoffman
- Department of Anatomy and Physiology, Nanotechnology Innovation Center (R.K.D., P.P., E.N.M., A.H.), Department of Anatomy and Physiology, Institute for Computational Comparative Medicine (Y.-H.C., Z.L.), and Department of Clinical Sciences (C.C., R.M.W., M.L.H.), College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Raelene M Wouda
- Department of Anatomy and Physiology, Nanotechnology Innovation Center (R.K.D., P.P., E.N.M., A.H.), Department of Anatomy and Physiology, Institute for Computational Comparative Medicine (Y.-H.C., Z.L.), and Department of Clinical Sciences (C.C., R.M.W., M.L.H.), College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Mary Lynn Higginbotham
- Department of Anatomy and Physiology, Nanotechnology Innovation Center (R.K.D., P.P., E.N.M., A.H.), Department of Anatomy and Physiology, Institute for Computational Comparative Medicine (Y.-H.C., Z.L.), and Department of Clinical Sciences (C.C., R.M.W., M.L.H.), College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| |
Collapse
|
80
|
Zinc oxide nanoparticles induce necroptosis and inhibit autophagy in MCF-7 human breast cancer cells. Biologia (Bratisl) 2019. [DOI: 10.2478/s11756-019-00325-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
81
|
Song WJ, Jeong MS, Choi DM, Kim KN, Wie MB. Zinc Oxide Nanoparticles Induce Autophagy and Apoptosis via Oxidative Injury and Pro-Inflammatory Cytokines in Primary Astrocyte Cultures. NANOMATERIALS 2019; 9:nano9071043. [PMID: 31330912 PMCID: PMC6669602 DOI: 10.3390/nano9071043] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/13/2019] [Accepted: 07/18/2019] [Indexed: 12/13/2022]
Abstract
The present study examined the potential toxic concentrations of zinc oxide nanoparticles (ZnO NPs) and associated autophagy and apoptosis-related injuries in primary neocortical astrocyte cultures. Concentrations of ZnO NPs ≥3 μg/mL induced significant toxicity in the astrocytes. At 24 h after exposure to the ZnO NPs, transmission electron microscopy revealed swelling of the endoplasmic reticulum (ER) and increased numbers of autophagolysosomes in the cultured astrocytes, and increased levels of LC3 (microtubule-associated protein 1 light chain 3)-mediated autophagy were identified by flow cytometry. Apoptosis induced by ZnO NP exposure was confirmed by the elevation of caspase-3/7 activity and 4′,6′-diamidino-2-phenylindole (DAPI) staining. Significant (p < 0.05) changes in the levels of glutathione peroxidase, superoxide dismutase, tumor necrosis factor (TNF-α), and interleukin-6 were observed by enzyme-linked immunoassay (ELISA) assay following the exposure of astrocyte cultures to ZnO NPs. Phosphatidylinositol 3-kinase (PI3K)/mitogen-activated protein kinase (MAPK) dual activation was induced by ZnO NPs in a dose-dependent manner. Additionally, the Akt (protein kinase B) inhibitor BML257 and the mTOR (mammalian target of rapamycin) inhibitor rapamycin contributed to the survival of astrocytes. Inhibitors of cyclooxygenase-2 and lipoxygenase attenuated ZnO NP-induced toxicity. Calcium-modulating compounds, antioxidants, and zinc/iron chelators also decreased ZnO NP-induced toxicity. Together, these results suggest that ZnO NP-induced autophagy and apoptosis may be associated with oxidative stress and the inflammatory process in primary astrocyte cultures.
Collapse
Affiliation(s)
- Woo-Ju Song
- Department of Veterinary Toxicology, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea
| | - Myung-Seon Jeong
- Chuncheon Center, Korean Basic Science Institute, Chuncheon 24341, Korea
- Department of Biochemistry, Kangwon National University, Chuncheon 24341, Korea
| | - Dong-Min Choi
- Department of Veterinary Toxicology, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea
| | - Kil-Nam Kim
- Chuncheon Center, Korean Basic Science Institute, Chuncheon 24341, Korea
| | - Myung-Bok Wie
- Department of Veterinary Toxicology, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Korea.
| |
Collapse
|
82
|
Yang Y, Cheng L, Deng X, Yu H, Chao L. Expression of GRIM-19 in unexplained recurrent spontaneous abortion and possible pathogenesis. Mol Hum Reprod 2019; 24:366-374. [PMID: 29741731 DOI: 10.1093/molehr/gay020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 05/07/2018] [Indexed: 12/16/2022] Open
Abstract
STUDY QUESTION Is aberrant expression of gene associated with retinoid-interferon-induced mortality-19 (GRIM-19) associated with unexplained recurrent spontaneous abortion (URSA)? SUMMARY ANSWER GRIM-19 deficiency may regulate regulatory T cell/T helper 17 cell (Treg/Th17) balance partly through reactive oxygen species (ROS)-mammalian target of rapamycin (mTOR) signaling axis in URSA. WHAT IS KNOWN ALREADY Immunological disorders may cause impaired maternal immune tolerance to the fetus and result in fetal rejection. The differentiation of Treg and Th17 cells is controlled by phosphoinositide 3-kinase (PI3K)/Akt/mTOR signaling pathway. GRIM-19 participates in the immune response, but its role in URSA is largely unknown. STUDY DESIGN, SIZE, DURATION The current study included 28 URSA patients and 30 non-pregnant healthy women. PARTICIPANTS/MATERIALS, SETTING, METHODS The proportion of Treg and Th17 cells in peripheral blood of URSA patients and control subjects were assessed with flow cytometry. The expression of GRIM-19 in peripheral blood lymphocytes (PBLs) was measured with quantitative real-time PCR and western blot analysis. Furthermore, the ROS level in the PBLs of URSA patients and control subjects were assessed by 2',7'-dichlorodihydrofluorescein diacetate (DCFH-DA) staining. Then, Akt/mTOR expression in the PBLs was measured. Downregulation of GRIM-19 in Jurkat cells was performed by specific siRNA. Then, intracellular ROS production and the expression of p-mTOR, which is known to enhance Th17 differentiation and decrease Treg cell differentiation, were detected. Finally, N-acetylcysteine (NAC) was used to decrease the intracellular ROS level, and the expression of p-mTOR was measured. MAIN RESULTS AND THE ROLE OF CHANCE The proportion of Treg cells was reduced in URSA patients, whereas the proportion of Th17 cells was increased. The expression of GRIM-19 was significantly lower in PBLs of URSA patients. Furthermore, there is a considerable increase in intracellular ROS production and a high level of p-Akt and p-mTOR expression in the PBLs of URSA patients compared with the control subjects. In parallel to this, downregulation of GRIM-19 in the Jurkat cells by siRNA results in an increased ROS production and an increased expression of p-mTOR. Importantly, the upregulation of p-mTOR resulting from GRIM-19 loss was significantly reversed in the cells treatment with ROS inhibitor N-acetyl-l-cysteine (NAC), indicating that ROS was indeed required for GRIM-19 depletion induced p-mTOR expression. LARGE SCALE DATA None. LIMITATIONS, REASONS FOR CAUTION A large number of researches have confirmed that the differentiation of Treg and Th17 cells is controlled by PI3K/Akt/mTOR signaling pathway. We have not shown the regulatory role of ROS and PI3K/Akt/mTOR in Treg and Th17 differentiation in this study. WIDER IMPLICATIONS OF THE FINDINGS Our study has demonstrated that GRIM-19 deficiency may play a role in regulating Treg/Th17 balance partly through ROS-mTOR signaling axis in URSA. The present study offers a new perspective to the roles of GRIM-19 in immunoregulation. STUDY FUNDING AND COMPETING INTEREST(S) This work was supported by the National Natural Science Foundation of China (Grant numbers 81571511, 81701528, 81370711 and 30901603), the Shandong Provincial Natural Science Foundation (Grant numbers ZR2017PH052 and ZR2013HM090) and the Science Foundation of Qilu Hospital of Shandong University, Fundamental Research Funds of Shandong University (Grant numbers 2015QLQN50 and 2015QLMS24). The authors declare that there is no conflict of interest that could prejudice the impartiality of the present research.
Collapse
Affiliation(s)
- Yang Yang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, PR China
| | - Laiyang Cheng
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, PR China
| | - Xiaohui Deng
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, PR China
| | - Hongling Yu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, PR China
| | - Lan Chao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, PR China
| |
Collapse
|
83
|
Yang H, Wang H, Liu Y, Yang L, Sun L, Tian Y, Zhao B, Lu H. The PI3K/Akt/mTOR signaling pathway plays a role in regulating aconitine-induced autophagy in mouse liver. Res Vet Sci 2019; 124:317-320. [PMID: 31030119 DOI: 10.1016/j.rvsc.2019.04.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 03/15/2019] [Accepted: 04/22/2019] [Indexed: 12/12/2022]
Abstract
Aconitine, a major aconitum alkaloid, is well known for its high toxicity that induces severe arrhythmias and neurological symptoms. One mechanism of aconitine-induced toxic responses is the induction of apoptosis. Apoptosis and autophagy are interconnected processes and the two pathways share critical components. In this study, we investigated the role of autophagy in aconitine-induced toxicity using mouse model. 120 mice were randomly divided into 4 experimental groups (normal saline), low dose group (0.14 μmol/L), medium dose group (0.28 μmol/L) and high dose group (0.56 μmol/ L). 30 mice in each group were administered with aconitine (lavage) for 30 days. The livers were collected for analysis of autophagy-related proteins by Western blotting. The expression of LC3II/LC3I ratio and Beclin 1 were found to increase and then decrease with the highest expression at 10 days and the p62 showed a time-dependent decreases. Autophagy is regulated by the mTOR pathway, we further analyzed the effects of aconitine on this pathway and found aconitine inhibited, phosphorylation of p-PI3K, p-Akt and p-mTOR. The p-p70s6k and p-4EBP1 which are downstream of mTOR were concomitantly decreased. These results suggest that aconitine induce autophagy in mouse liver. The PI3K/Akt/mTOR signaling pathway is involved in the regulation of aconitine-induced autophagy in the liver of mice.
Collapse
Affiliation(s)
- Hanqi Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Hui Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yanbing Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Lin Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Lu Sun
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yanan Tian
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA
| | - Baoyu Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Hao Lu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
84
|
Li X, Xu H, Li C, Qiao G, Farooqi AA, Gedanken A, Liu X, Lin X. Zinc-Doped Copper Oxide Nanocomposites Inhibit the Growth of Pancreatic Cancer by Inducing Autophagy Through AMPK/mTOR Pathway. Front Pharmacol 2019; 10:319. [PMID: 31001120 PMCID: PMC6454023 DOI: 10.3389/fphar.2019.00319] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/15/2019] [Indexed: 12/19/2022] Open
Abstract
Zinc doped copper oxide nanocomposites (Zn-CuO NPs) is a novel doped metal nanomaterial synthesized by our group using the sonochemical method. Our previous studies have shown that Zn-CuO NPs could inhibit cancer cell proliferation by inducing apoptosis via ROS-mediated pathway. In the present study, we studied the anticancer effect of Zn-CuO NPs on human pancreatic cancer cells. MTS assay revealed that Zn-CuO NPs was able to inhibit cancer cell growth. TEM, flow cytometry and fluorescence microscope analysis showed that Zn-CuO NPs induced autophagy significantly; the number of autophagosomes increased obviously in cells treated with Zn-CuO NPs. Western blot analysis revealed that treatment with the NPs resulted in activation of AMPK/mTOR pathway in both AsPC-1 and MIA Paca-2 cells in dose dependent manners. Moreover, in the presence of AMPK activator AMPKinone, the protein level of p-AMPK, p-ULK1, Beclin-1 and LC3-II/LC3-I increased, while the protein expression of p-AMPK, p-ULK1, Beclin-1 and LC3-II/LC3-I decreased in the presence of AMPK inhibitor Compound C. In vivo study using xenograft mice revealed that Zn-CuO NPs significantly inhibited tumor growth with low toxicity. Our study confirms that Zn-CuO NPs inhibit the tumor growth both in vitro and in vivo for pancreatic cancer. AMPK/mTOR pathway plays an important role in the NPs induced inhibition of tumor growth.
Collapse
Affiliation(s)
- Xiao Li
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Huanli Xu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Cong Li
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Gan Qiao
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ammad Ahmad Farooqi
- Laboratory for Translational Oncology and Personalized Medicine, Rashid Latif Medical College (RLMC), Lahore, Pakistan
| | - Aharon Gedanken
- Center for Advanced Materials and Nanotechnology, Bar-Ilan University, Ramat Gan, Israel
| | - Xiaohui Liu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiukun Lin
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
85
|
Augustine R, Prasad P, Khalaf IMN. Therapeutic angiogenesis: From conventional approaches to recent nanotechnology-based interventions. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 97:994-1008. [DOI: 10.1016/j.msec.2019.01.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 12/06/2018] [Accepted: 01/02/2019] [Indexed: 12/27/2022]
|
86
|
Cordani M, Somoza Á. Targeting autophagy using metallic nanoparticles: a promising strategy for cancer treatment. Cell Mol Life Sci 2019; 76:1215-1242. [PMID: 30483817 PMCID: PMC6420884 DOI: 10.1007/s00018-018-2973-y] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 11/20/2018] [Indexed: 02/08/2023]
Abstract
Despite the extensive genetic and phenotypic variations present in the different tumors, they frequently share common metabolic alterations, such as autophagy. Autophagy is a self-degradative process in response to stresses by which damaged macromolecules and organelles are targeted by autophagic vesicles to lysosomes and then eliminated. It is known that autophagy dysfunctions can promote tumorigenesis and cancer development, but, interestingly, its overstimulation by cytotoxic drugs may also induce cell death and chemosensitivity. For this reason, the possibility to modulate autophagy may represent a valid therapeutic approach to treat different types of cancers and a variety of clinical trials, using autophagy modulators, are currently employed. On the other hand, recent progress in nanotechnology offers plenty of tools to fight cancer with innovative and efficient therapeutic agents by overcoming obstacles usually encountered with traditional drugs. Interestingly, nanomaterials can modulate autophagy and have been exploited as therapeutic agents against cancer. In this article, we summarize the most recent advances in the application of metallic nanostructures as potent modulators of autophagy process through multiple mechanisms, stressing their therapeutic implications in cancer diseases. For this reason, we believe that autophagy modulation with nanoparticle-based strategies would acquire clinical relevance in the near future, as a complementary therapy for the treatment of cancers and other diseases.
Collapse
Affiliation(s)
- Marco Cordani
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), CNB-CSIC-IMDEA Nanociencia Associated Unit "Unidad de Nanobiotecnología", Madrid, Spain.
- Institute for Advanced Studies in Nanoscience (IMDEA Nanociencia), Faraday 9, Office 129, Lab 137 Ciudad Universitaria de Cantoblanco, 28049, Madrid, Spain.
| | - Álvaro Somoza
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), CNB-CSIC-IMDEA Nanociencia Associated Unit "Unidad de Nanobiotecnología", Madrid, Spain.
- Institute for Advanced Studies in Nanoscience (IMDEA Nanociencia), Faraday 9, Office 129, Lab 137 Ciudad Universitaria de Cantoblanco, 28049, Madrid, Spain.
| |
Collapse
|
87
|
Hassan I, Husain FM, Khan RA, Ebaid H, Al-Tamimi J, Alhazza IM, Aman S, Ibrahim KE. Ameliorative effect of zinc oxide nanoparticles against potassium bromate-mediated toxicity in Swiss albino rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:9966-9980. [PMID: 30739294 DOI: 10.1007/s11356-019-04443-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 01/30/2019] [Indexed: 06/09/2023]
Abstract
Potassium bromate (PB) is a commonly used food additive, a prominent water disinfection by-product, and a class IIB carcinogen. It exerts a various degree of toxicity depending on its dose and exposure duration consumed with food and water in the living organisms. The present investigation aims to demonstrate the protective efficacy of zinc oxide nanoparticles (ZnO NPs) derived from Ochradenus arabicus (OA) leaf extract by green technology in PB-challenged Swiss albino rats. The rodents were randomly distributed, under the lab-standardized treatment strategy, into the following six treatment groups: control (group I), PB alone (group II), ZnO alone (group III), ZnO NP alone (group IV), PB + ZnO (group V), and PB + ZnO NPs (group VI). The rats were sacrificed after completion of the treatment, and their blood and liver samples were collected for further analysis. Group II showed extensive toxic effects with altered liver function markers (alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, lactate dehydrogenase, gamma-glutamyl transferase, glutathione-S-transferase, and thioredoxin reductase) and compromised redox status (SOD, CAT, GR, GPx, GSH, MDA, and total carbonyl content). The histopathological analysis and comet assay further supported the biochemical results of the same group. Besides, group III also showed moderate toxicity evidenced by an alteration in most of the studied parameters while group IV demonstrated mild toxicity after biochemical analysis indicating the excellent biocompatibility of the NPs. However, group VI exhibited attenuation of the PB-induced toxic insults to a significant level as compared to group II, whereas group V failed to show similar improvement in the studied parameters. All these findings entail that the ZnO NPs prepared by green synthesis have significant ameliorative property against PB-induced toxicity in vivo. Moreover, administration of the NPs improved the overall health of the treated animals profoundly. Hence, these NPs have significant therapeutic potential against the toxic effects of PB and similar compounds in vivo, and they are suitable to be used at the clinical and industrial levels.
Collapse
Affiliation(s)
- Iftekhar Hassan
- Department of Zoology, College of Science, King Saud University, Building 05, Riyadh, 11451, Saudi Arabia.
| | - Fohad Mabood Husain
- Department of Food Science and Nutrition, College of Food and Agriculture, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Rais Ahmad Khan
- Department of Chemistry, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Hossam Ebaid
- Department of Zoology, College of Science, King Saud University, Building 05, Riyadh, 11451, Saudi Arabia
| | - Jameel Al-Tamimi
- Department of Zoology, College of Science, King Saud University, Building 05, Riyadh, 11451, Saudi Arabia
| | - Ibrahim M Alhazza
- Department of Zoology, College of Science, King Saud University, Building 05, Riyadh, 11451, Saudi Arabia
| | - Shazia Aman
- Department of Biochemistry, J N Medical College and Hospital, Aligarh Muslim University, Aligarh, 202002, India
| | - Khalid Elfaki Ibrahim
- Department of Zoology, College of Science, King Saud University, Building 05, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
88
|
Lunova M, Smolková B, Lynnyk A, Uzhytchak M, Jirsa M, Kubinová Š, Dejneka A, Lunov O. Targeting the mTOR Signaling Pathway Utilizing Nanoparticles: A Critical Overview. Cancers (Basel) 2019; 11:E82. [PMID: 30642006 PMCID: PMC6356373 DOI: 10.3390/cancers11010082] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/21/2018] [Accepted: 01/05/2019] [Indexed: 12/21/2022] Open
Abstract
Proteins of the mammalian target of rapamycin (mTOR) signaling axis are overexpressed or mutated in cancers. However, clinical inhibition of mTOR signaling as a therapeutic strategy in oncology shows rather limited progress. Nanoparticle-based mTOR targeted therapy proposes an attractive therapeutic option for various types of cancers. Along with the progress in the biomedical applications of nanoparticles, we start to realize the challenges and opportunities that lie ahead. Here, we critically analyze the current literature on the modulation of mTOR activity by nanoparticles, demonstrate the complexity of cellular responses to functionalized nanoparticles, and underline challenges lying in the identification of the molecular mechanisms of mTOR signaling affected by nanoparticles. We propose the idea that subcytotoxic doses of nanoparticles could be relevant for the induction of subcellular structural changes with possible involvement of mTORC1 signaling. The evaluation of the mechanisms and therapeutic effects of nanoparticle-based mTOR modulation will provide fundamental knowledge which could help in developing safe and efficient nano-therapeutics.
Collapse
Affiliation(s)
- Mariia Lunova
- Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic.
- Institute for Clinical & Experimental Medicine (IKEM), Prague, 140 21, Czech Republic.
| | - Barbora Smolková
- Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic.
| | - Anna Lynnyk
- Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic.
| | - Mariia Uzhytchak
- Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic.
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), Prague, 140 21, Czech Republic.
| | - Šárka Kubinová
- Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic.
- Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, 14220, Czech Republic.
| | - Alexandr Dejneka
- Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic.
| | - Oleg Lunov
- Institute of Physics of the Czech Academy of Sciences, Prague, 18221, Czech Republic.
| |
Collapse
|
89
|
Mohammadinejad R, Moosavi MA, Tavakol S, Vardar DÖ, Hosseini A, Rahmati M, Dini L, Hussain S, Mandegary A, Klionsky DJ. Necrotic, apoptotic and autophagic cell fates triggered by nanoparticles. Autophagy 2019; 15:4-33. [PMID: 30160607 PMCID: PMC6287681 DOI: 10.1080/15548627.2018.1509171] [Citation(s) in RCA: 271] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 07/19/2018] [Accepted: 08/03/2018] [Indexed: 12/15/2022] Open
Abstract
Nanomaterials have gained a rapid increase in use in a variety of applications that pertain to many aspects of human life. The majority of these innovations are centered on medical applications and a range of industrial and environmental uses ranging from electronics to environmental remediation. Despite the advantages of NPs, the knowledge of their toxicological behavior and their interactions with the cellular machinery that determines cell fate is extremely limited. This review is an attempt to summarize and increase our understanding of the mechanistic basis of nanomaterial interactions with the cellular machinery that governs cell fate and activity. We review the mechanisms of NP-induced necrosis, apoptosis and autophagy and potential implications of these pathways in nanomaterial-induced outcomes. Abbreviations: Ag, silver; CdTe, cadmium telluride; CNTs, carbon nanotubes; EC, endothelial cell; GFP, green fluorescent protein; GO, graphene oxide; GSH, glutathione; HUVECs, human umbilical vein endothelial cells; NP, nanoparticle; PEI, polyethylenimine; PVP, polyvinylpyrrolidone; QD, quantum dot; ROS, reactive oxygen species; SiO2, silicon dioxide; SPIONs, superparamagnetic iron oxide nanoparticles; SWCNT, single-walled carbon nanotubes; TiO2, titanium dioxide; USPION, ultra-small super paramagnetic iron oxide; ZnO, zinc oxide.
Collapse
Affiliation(s)
- Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Amin Moosavi
- Department of Molecular Medicine, Institute of Medical Biotechnology, National Institute for Genetic Engineering and Biotechnology, Tehran, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Deniz Özkan Vardar
- Sungurlu Vocational High School, Health Programs, Hitit University, Corum, Turkey
| | - Asieh Hosseini
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Marveh Rahmati
- Cancer Biology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Salik Hussain
- Department of Physiology, Pharmacology and Neuroscience, West Virginia University, School of Medicine, Morgantown, WV, USA
| | - Ali Mandegary
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | | |
Collapse
|
90
|
Wu N, Zhang C, Wang C, Song L, Yao W, Gedanken A, Lin X, Shi D. Zinc-doped copper oxide nanocomposites reverse temozolomide resistance in glioblastoma by inhibiting AKT and ERK1/2. Nanomedicine (Lond) 2018; 13:1303-1318. [PMID: 29949469 DOI: 10.2217/nnm-2017-0359] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AIM To assess the effect of zinc-doped copper oxide nanocomposites (nZn-CuO NPs) on glioblastoma therapy. MATERIALS & METHODS nZn-CuO NPs were synthesized by sonochemical method and its antitumor effects and underlying molecular mechanisms were investigated both in vitro and in vivo. RESULTS After nZn-CuO NPs treatment, cell proliferation was significantly inhibited in dividing cancer cells but less toxicity was observed in normal cells. In vivo studies show that nZn-CuO NPs inhibited tumor growth in a dose-dependent manner. Further study found that nZn-CuO NPs trigger cell reactive oxygen species (ROS) generation and intrinsic apoptotic pathway. In temozolomide resistance glioblastoma, nZn-CuO NPs disturb cell growth and sphere formation by inhibiting AKT and ERK1/2 activation. CONCLUSION nZn-CuO NPs possess the potential to be developed as a novel anti-tumor agent, especially to treat temozolomide resistance glioblastoma.
Collapse
Affiliation(s)
- Ning Wu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Drugs & Bioproducts, Qingdao National Laboratory for Marine Science & Technology, Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Chunyun Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Qingdao, China
| | - Changhui Wang
- Shanghai Neuromedical Center, Qingdao University, Shanghai, China
| | - Lairong Song
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Weicheng Yao
- Department of Neurosurgery, Qingdao University, Qingdao, China
| | - Aharon Gedanken
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Xiukun Lin
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Dayong Shi
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Drugs & Bioproducts, Qingdao National Laboratory for Marine Science & Technology, Qingdao, China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| |
Collapse
|
91
|
The Role of Zinc and Zinc Homeostasis in Macrophage Function. J Immunol Res 2018; 2018:6872621. [PMID: 30622979 PMCID: PMC6304900 DOI: 10.1155/2018/6872621] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/31/2018] [Accepted: 11/06/2018] [Indexed: 01/12/2023] Open
Abstract
Zinc has long been recognized as an essential trace element, playing roles in the growth and development of all living organisms. In recent decades, zinc homeostasis was also found to be important for the innate immune system, especially for maintaining the function of macrophages. It is now generally accepted that dysregulated zinc homeostasis in macrophages causes impaired phagocytosis and an abnormal inflammatory response. However, many questions remain with respect to the mechanisms that underlie these processes, particularly at the cellular and molecular levels. Here, we review our current understanding of the roles that zinc and zinc transporters play in regulating macrophage function.
Collapse
|
92
|
Ye M, Shi B. Zirconia Nanoparticles-Induced Toxic Effects in Osteoblast-Like 3T3-E1 Cells. NANOSCALE RESEARCH LETTERS 2018; 13:353. [PMID: 30402719 PMCID: PMC6219995 DOI: 10.1186/s11671-018-2747-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 10/11/2018] [Indexed: 05/27/2023]
Abstract
Zirconia (ZrO2) is one of the widely used metal oxides for potential bio-applications such as biosensors, cancer therapy, implants, and dentistry due to its high mechanical strength and less toxicity. Because of their widespread applications, the potential exposure to these nanoparticles (NPs) has increased, which has attracted extensive attention. Thus, it is urgent to investigate the toxicological profile of ZrO2 NPs. Titanium dioxide (TiO2) is another extensively used nanomaterials which are known to be weakly toxic. In this study, TiO2 NPs were served as control to evaluate the biocompatibility of ZrO2 NPs. We detected the cytotoxicity of TiO2 and ZrO2 NPs in osteoblast-like 3T3-E1 cells and found that reactive oxygen species (ROS) played a crucial role in the TiO2 and ZrO2 NP-induced cytotoxicity with concentration-dependent manner. We also showed TiO2 and ZrO2 NPs could induce apoptosis and morphology changes after culturing with 3T3-E1 cells at high concentrations. Moreover, TiO2 and ZrO2 NPs at high concentrations could inhibit cell osteogenic differentiation, compared to those at low concentrations. In conclusion, TiO2 and ZrO2 NPs could induce cytotoxic responses in vitro in a concentration-dependent manner, which may also affect osteogenesis; ZrO2 NPs showed more potent toxic effects than TiO2 NPs.
Collapse
Affiliation(s)
- Mingfu Ye
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedical Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, People's Republic of China
- Department of Implantology, Xiamen Stomatology Hospital, Hospital and School of Stomatology, Xiamen Medical University, Xiamen, 361003, People's Republic of China
| | - Bin Shi
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedical Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, People's Republic of China.
- Department of Implantology, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Rd, Wuhan, 430072, People's Republic of China.
| |
Collapse
|
93
|
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
|
94
|
Jiang X, Tang Q, Zhang J, Wang H, Bai L, Meng P, Qin X, Xu G, Bose DD, Wang B, Chen C, Zou Z. Autophagy-dependent release of zinc ions is critical for acute lung injury triggered by zinc oxide nanoparticles. Nanotoxicology 2018; 12:1068-1091. [PMID: 30317896 DOI: 10.1080/17435390.2018.1513094] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Pulmonary exposure to zinc oxide nanoparticles (ZnONPs) could cause acute lung injury (ALI), but the underlying molecular mechanism remains unclear. Herein, we established a ZnONPs-induced ALI mouse model, characterized by the histopathological changes (edema and infiltration of inflammatory cells in lung tissues), and the elevation of total protein and cytokine interleukin-6 in bronchoalveolar lavage fluid in time- and dose-dependent manners. This model also exhibited features like the disturbance of redox-state (reduced of glutathione to glutathione disulfide ratio, elevation of heme oxygenase-1 and superoxide dismutase 2), the decrease of adenosine triphosphate synthesis and the release of zinc ions in the lung tissues. Interestingly, we found that ZnONPs exposure caused the accumulation of autophagic vacuoles and the elevation of microtubule-associated proteins 1A/1B light chain (LC)3B-II and p62, indicating the impairment of autophagic flux. Our data indicated that the above process might be regulated by the activation of AMP-activated protein kinase but not the mammalian target of rapamycin pathway. The association between ZnONPs-induced ALI and autophagy was further verified by a classical autophagy inhibitor, 3-methyladenine (3-MA). 3-MA administration reduced the accumulation of autophagic vacuoles, the expression of LC3B-II and p62, followed by a significant attenuation of histopathological changes, inflammation, and oxidative stress. More importantly, 3-MA could directly decrease the release of zinc ions in lung tissues. Taken together, our study provides the evidence that ZnONPs-induced pulmonary toxicity is autophagy-dependent, suggests that limiting the release of zinc ions by inhibiting autophagy could be a feasible strategy for the prevention of ZnONPs-associated pulmonary toxicity.
Collapse
Affiliation(s)
- Xuejun Jiang
- a Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center , Chongqing Medical University , Chongqing , People's Republic of China.,b Laboratory of Tissue and Cell Biology, Experimental Teaching and Management Center , Chongqing Medical University , Chongqing , People's Republic of China
| | - Qianghu Tang
- c Department of Occupational and Environmental Health, School of Public Health and Management , Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University , Chongqing , People's Republic of China
| | - Jun Zhang
- d Institute of Life Sciences, Chongqing Medical University , Chongqing , People's Republic of China
| | - Hong Wang
- d Institute of Life Sciences, Chongqing Medical University , Chongqing , People's Republic of China
| | - Lulu Bai
- c Department of Occupational and Environmental Health, School of Public Health and Management , Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University , Chongqing , People's Republic of China
| | - Pan Meng
- c Department of Occupational and Environmental Health, School of Public Health and Management , Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University , Chongqing , People's Republic of China
| | - Xia Qin
- e Department of Pharmacy , The First Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Ge Xu
- d Institute of Life Sciences, Chongqing Medical University , Chongqing , People's Republic of China
| | - Diptiman D Bose
- f Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences , Western New England University , Springfield , MA , USA
| | - Bin Wang
- d Institute of Life Sciences, Chongqing Medical University , Chongqing , People's Republic of China
| | - Chengzhi Chen
- c Department of Occupational and Environmental Health, School of Public Health and Management , Research Center for Medicine and Social Development, Innovation Center for Social Risk Governance in Health, Chongqing Medical University , Chongqing , People's Republic of China.,g Post-doctoral Research Stations of Nursing Science, School of Nursing , Chongqing Medical University , Chongqing , People's Republic of China
| | - Zhen Zou
- d Institute of Life Sciences, Chongqing Medical University , Chongqing , People's Republic of China
| |
Collapse
|
95
|
He G, Ma Y, Zhu Y, Yong L, Liu X, Wang P, Liang C, Yang C, Zhao Z, Hai B, Pan X, Liu Z, Liu X, Mao C. Cross Talk Between Autophagy and Apoptosis Contributes to ZnO Nanoparticle-Induced Human Osteosarcoma Cell Death. Adv Healthc Mater 2018; 7:e1800332. [PMID: 29900694 PMCID: PMC6310009 DOI: 10.1002/adhm.201800332] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/18/2018] [Indexed: 12/13/2022]
Abstract
Killing osteosarcoma cells by zinc oxide nanoparticles (NPs) and its underlying subcellular mechanism are never studied. Here, it is found that the NPs induce cross talk between apoptosis and autophagy, which leads to osteosarcoma cell death. Specifically, the NP uptake promotes autophagy by inducing accumulation of autophagosomes along with impairment of lysosomal functions. The autophagy further causes the uptaken NPs to release zinc ions by promoting their dissolution. These intracellular zinc ions, together with those that are originally released from the extracellular NPs and flowed into the cells, collectively target and damage mitochondria to produce reactive oxygen species (ROS). Then the ROS inhibit cell proliferation by arresting S phase and trigger apoptosis by extrinsic and intrinsic pathways, ultimately leading to cell death. More importantly, suppression of the early stage autophagy restores cell viability by abolishing apoptosis whereas blockade of the late stage autophagy inversely enhances apoptosis. In contrast, inhibition of apoptosis shows a limited ability to restore cell viability but obviously enhance autophagy. Notably, cell viability is strongly ameliorated by the combination of inhibitors for both the late stage autophagy and the apoptosis. These findings provide a mechanistic understanding of the NP-directed autophagy and apoptosis in osteosarcoma cells.
Collapse
Affiliation(s)
- Guanping He
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Yunlong Ma
- The Center for Pain Medicine, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Ye Zhu
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Lei Yong
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Xiao Liu
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Peng Wang
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Chen Liang
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Chenlong Yang
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Zhigang Zhao
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Bao Hai
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Xiaoyu Pan
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Zhongjun Liu
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Xiaoguang Liu
- Department of Orthopedics, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, USA
| |
Collapse
|
96
|
Li Y, Ju D. The Role of Autophagy in Nanoparticles-Induced Toxicity and Its Related Cellular and Molecular Mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1048:71-84. [PMID: 29453533 DOI: 10.1007/978-3-319-72041-8_5] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the past decades, nanoparticles have been widely used in industry and pharmaceutical fields for drug delivery, anti-pathogen, and diagnostic imaging purposes because of their unique physicochemical characteristics such as special ultrastructure, dispersity, and effective cellular uptake properties. But the nanotoxicity has been raised over the extensive applications of nanoparticles. Researchers have elucidated series of mechanisms in nanoparticles-induced toxicity, including apoptosis, necrosis, oxidative stress, and autophagy. Among upon mechanisms, autophagy was recently recognized as an important cell death style in various nanoparticles-induced toxicity, but the role of autophagy and its related cellular and molecular mechanisms during nanoparticles-triggered toxicity were still confusing. In the chapter, we briefly introduced the general process of autophagy, summarized the different roles of autophagy in various nanoparticle-treated different in vitro/in vivo models, and deeply analyzed the physicochemical and biochemical (cellular and molecular) mechanisms of autophagy during nanoparticles-induced toxicity through listing and summarizing representative examples. Physicochemical mechanisms mainly include dispersity, size, charge, and surface chemistry; cellular mechanisms primarily focus on lysosome impairment, mitochondria dysfunction, mitophagy, endoplasmic reticulum stress and endoplasmic reticulum autophagy; while molecular mechanisms were mainly including autophagy related signaling pathways, hypoxia-inducible factor, and oxidative stress. This chapter highlighted the important role of autophagy as a critical mechanism in nanoparticles-induced toxicity, and the physicochemical and biochemical mechanisms of autophagy triggered by nanoparticles might be useful for establishing a guideline for the evaluation of nanotoxicology, designing and developing new biosafety nanoparticles in the future.
Collapse
Affiliation(s)
- Yubin Li
- Department of Microbiological and Biochemical Pharmacy & The Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, People's Republic of China. .,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Dianwen Ju
- Department of Microbiological and Biochemical Pharmacy & The Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
97
|
Causation by Diesel Exhaust Particles of Endothelial Dysfunctions in Cytotoxicity, Pro-inflammation, Permeability, and Apoptosis Induced by ROS Generation. Cardiovasc Toxicol 2018; 17:384-392. [PMID: 26965709 DOI: 10.1007/s12012-016-9364-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Epidemiological studies suggest that an increase of diesel exhaust particles (DEP) in ambient air corresponds to an increase in hospital-recorded myocardial infarctions within 48 h after exposure. Among the many theories to explain this data are endothelial dysfunction and translocation of DEP into vasculature. The mechanisms for such DEP-induced vascular permeability remain unknown. One of the major mechanisms underlying the effects of DEP is suggested to be oxidative stress. Experiments have shown that DEP induce the generation of reactive oxygen species (ROS), such as superoxide anion and H2O2 in the HUVEC tube cells. Transcription factor Nrf2 is translocated to the cell nucleus, where it activates transcription of the antioxidative enzyme HO-1 and sequentially induces the release of vascular permeability factor VEGF-A. Furthermore, a recent study shows that DEP-induced intracellular ROS may cause the release of pro-inflammatory TNF-α and IL-6, which may induce endothelial permeability as well by promoting VEGF-A secretion independently of HO-1 activation. These results demonstrated that the adherens junction molecule, VE-cadherin, becomes redistributed from the membrane at cell-cell borders to the cytoplasm in response to DEP, separating the plasma membranes of adjacent cells. DEP were occasionally found in endothelial cell cytoplasm and in tube lumen. In addition, the induced ROS is cytotoxic to the endothelial tube-like HUVEC. Acute DEP exposure stimulates ATP depletion, followed by depolarization of their actin cytoskeleton, which sequentially inhibits PI3K/Akt activity and induces endothelial apoptosis. Nevertheless, high-dose DEP augments tube cell apoptosis up to 70 % but disrupts the p53 negative regulator Mdm2. In summary, exposure to DEP affects parameters influencing vasculature permeability and viability, i.e., oxidative stress and its upregulated antioxidative and pro-inflammatory responses, which sequentially induce vascular permeability factor, VEGF-A release and disrupt cell-cell junction integrity. While exposure to a low dose of DEP actin triggers cytoskeleton depolarization, reduces PI3K/Akt activity, and induces a p53/Mdm2 feedback loop, a high dose causes apoptosis by depleting Mdm2. Addition of ROS scavenger N-acetyl cysteine suppresses DEP-induced oxidative stress efficiently and reduces subsequent damages by increasing endogenous glutathione.
Collapse
|
98
|
Chen QY, Costa M. PI3K/Akt/mTOR Signaling Pathway and the Biphasic Effect of Arsenic in Carcinogenesis. Mol Pharmacol 2018; 94:784-792. [PMID: 29769245 PMCID: PMC5994485 DOI: 10.1124/mol.118.112268] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/30/2018] [Indexed: 12/22/2022] Open
Abstract
Arsenic is a naturally occurring, ubiquitous metalloid found in the Earth's crust. In its inorganic form, arsenic is highly toxic and carcinogenic and is widely found across the globe and throughout the environment. As an International Agency for Research on Cancer-defined class 1 human carcinogen, arsenic can cause multiple human cancers, including liver, lung, urinary bladder, skin, kidney, and prostate. Mechanisms of arsenic-induced carcinogenesis remain elusive, and this review focuses specifically on the role of the PI3K/AKT/mTOR pathway in promoting cancer development. In addition to exerting potent carcinogenic responses, arsenic is also known for its therapeutic effects against acute promyelocytic leukemia. Current literature suggests that arsenic can achieve both therapeutic as well as carcinogenic effects, and this review serves to examine the paradoxical effects of arsenic, specifically through the PI3K/AKT/mTOR pathway. Furthermore, a comprehensive review of current literature reveals an imperative need for future studies to establish and pinpoint the exact conditions for which arsenic can, and through what mechanisms it is able to, differentially regulate the PI3K/AKT/mTOR pathway to maximize the therapeutic and minimize the carcinogenic properties of arsenic.
Collapse
Affiliation(s)
- Qiao Yi Chen
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York
| | - Max Costa
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York
| |
Collapse
|
99
|
Inhibitory effects of selenium on cadmium-induced cytotoxicity in PC12 cells via regulating oxidative stress and apoptosis. Food Chem Toxicol 2018; 114:180-189. [DOI: 10.1016/j.fct.2018.02.034] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 12/30/2022]
|
100
|
Pellacani C, Costa LG. Role of autophagy in environmental neurotoxicity. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 235:791-805. [PMID: 29353798 DOI: 10.1016/j.envpol.2017.12.102] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 12/08/2017] [Accepted: 12/25/2017] [Indexed: 06/07/2023]
Abstract
Human exposure to neurotoxic pollutants (e.g. metals, pesticides and other chemicals) is recognized as a key risk factor in the pathogenesis of neurodegenerative disorders. Emerging evidence indicates that an alteration in autophagic pathways may be correlated with the onset of the neurotoxicity resulting from chronic exposure to these pollutants. In fact, autophagy is a natural process that permits to preserving cell homeostasis, through the seizure and degradation of the cytosolic damaged elements. However, when an excessive level of intracellular damage is reached, the autophagic process may also induce cell death. A correct modulation of specific stages of autophagy is important to maintain the correct balance in the organism. In this review, we highlight the critical role that autophagy plays in neurotoxicity induced by the most common classes of environmental contaminants. The understanding of this mechanism may be helpful to discover a potential therapeutic strategy to reduce side effects induced by these compounds.
Collapse
Affiliation(s)
- C Pellacani
- Dept. of Medicine and Surgery, University of Parma, Parma, Italy.
| | - L G Costa
- Dept. of Medicine and Surgery, University of Parma, Parma, Italy; Dept. of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| |
Collapse
|