1
|
Lam W, Yao Y, Tang C, Wang Y, Yuan Q, Peng L. Bifunctional mesoporous HMUiO-66-NH 2 nanoparticles for bone remodeling and ROS scavenging in periodontitis therapy. Biomaterials 2025; 314:122872. [PMID: 39383779 DOI: 10.1016/j.biomaterials.2024.122872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 09/17/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Periodontal bone defects represent an irreversible consequence of periodontitis associated with reactive oxygen species (ROS). However, indiscriminate removal of ROS proves to be counterproductive for tissue repair and insufficient for addressing existing bone defects. In the treatment of periodontitis, it is crucial to rationally alleviate local ROS while simultaneously promoting bone regeneration. In this study, Zr-based large-pore hierarchical mesoporous metal-organic framework (MOF) nanoparticles (NPs) HMUiO-66-NH2 were successfully proposed as bifunctional nanomaterials for bone regeneration and ROS scavenging in periodontitis therapy. HMUiO-66-NH2 NPs demonstrated outstanding biocompatibility both in vitro and in vivo. Significantly, these NPs enhanced the osteogenic differentiation of bone mesenchymal stem cells (BMSCs) under normal and high ROS conditions, upregulating osteogenic gene expression and mitigating oxidative stress. Furthermore, in vivo imaging revealed a gradual degradation of HMUiO-66-NH2 NPs in periodontal tissues. Local injection of HMUiO-66-NH2 effectively reduced bone defects and ROS levels in periodontitis-induced C57BL/6 mice. RNA sequencing highlighted that differentially expressed genes (DEGs) are predominantly involved in bone tissue development, with notable upregulation in Wnt and TGF-β signaling pathways. In conclusion, HMUiO-66-NH2 exhibits dual functionality in alleviating oxidative stress and promoting bone repair, positioning it as an effective strategy against bone resorption in oxidative stress-related periodontitis.
Collapse
Affiliation(s)
- Waishan Lam
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yufei Yao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, 610041, China
| | - Chenxi Tang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yue Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral Surgery, West China Hospital of Stomatology, Sichuan University, Sichuan, 610041, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lin Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
2
|
Tsourmas KI, Butler CA, Kwang NE, Sloane ZR, Dykman KJG, Maloof GO, Prekopa CA, Krattli RP, El-Khatib SM, Swarup V, Acharya MM, Hohsfield LA, Green KN. Myeloid-derived β-hexosaminidase is essential for neuronal health and lysosome function: implications for Sandhoff disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.21.619538. [PMID: 39484433 PMCID: PMC11526954 DOI: 10.1101/2024.10.21.619538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Lysosomal storage disorders (LSDs) are a large disease class involving lysosomal dysfunction, often resulting in neurodegeneration. Sandhoff disease (SD) is an LSD caused by a deficiency in the β subunit of the β-hexosaminidase enzyme (Hexb). Although Hexb expression in the brain is specific to microglia, SD primarily affects neurons. To understand how a microglial gene is involved in maintaining neuronal homeostasis, we demonstrated that β-hexosaminidase is secreted by microglia and integrated into the neuronal lysosomal compartment. To assess therapeutic relevance, we treated SD mice with bone marrow transplant and colony stimulating factor 1 receptor inhibition, which broadly replaced Hexb -/- microglia with Hexb-sufficient cells. This intervention reversed apoptotic gene signatures, improved behavior, restored enzymatic activity and Hexb expression, ameliorated substrate accumulation, and normalized neuronal lysosomal phenotypes. These results underscore the critical role of myeloid-derived β-hexosaminidase in neuronal lysosomal function and establish microglial replacement as a potential LSD therapy.
Collapse
Affiliation(s)
- Kate I. Tsourmas
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Claire A. Butler
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Nellie E. Kwang
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Zachary R. Sloane
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Koby J. G. Dykman
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Ghassan O. Maloof
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Christiana A. Prekopa
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Robert P. Krattli
- Department of Anatomy and Neurobiology; University of California; Irvine, CA 92697; USA
| | - Sanad M. El-Khatib
- Department of Anatomy and Neurobiology; University of California; Irvine, CA 92697; USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Munjal M. Acharya
- Department of Anatomy and Neurobiology; University of California; Irvine, CA 92697; USA
- Department of Radiation Oncology; University of California; Irvine, CA 92697; USA
| | - Lindsay A. Hohsfield
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| | - Kim N. Green
- Department of Neurobiology and Behavior; University of California; Irvine, CA 92697; USA
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine, CA 92697; USA
| |
Collapse
|
3
|
Chen X, Bao Y, Sun G, Wang X, Zhu J. UNC13B regulates the sensitivity of Wilms' tumor cells to doxorubicin by modulating lysosomes. Oncol Lett 2024; 28:446. [PMID: 39091580 PMCID: PMC11292464 DOI: 10.3892/ol.2024.14579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 06/14/2024] [Indexed: 08/04/2024] Open
Abstract
Wilms' tumor is a malignant neoplasm where current medical advancements have significantly improved survival rates; however, challenges persist such as the resistance of the tumor to chemotherapy drugs like doxorubicin. This necessitates higher dosages, leading to decreased sensitivity. However, using high doses of doxorubicin can have late effects on the heart. Unc-13 homolog B (UNC13B) may be involved in the drug resistance in several tumors, yet its role in modulating drug sensitivity in Wilms' tumor remains unexplored. UNC13B levels were quantified using reverse transcription-qPCR and Western blotting. The half-maximal inhibitory concentration for doxorubicin, vincristine, and actinomycin-D was determined using CCK-8 assays. Cell cycle and apoptosis were analyzed using flow cytometry, and lysosomal changes were observed using Lyso-Tracker staining. The present study initially evaluated UNC13B expression levels in the Wilms' tumor 17.94 cell line. Additionally, through short hairpin RNA-mediated knockdown, changes in doxorubicin sensitivity in 17.94 Wilms' tumor cells were assessed. Concurrently, preliminary investigations into the role of UNC13B in regulating lysosomes was performed, revealing a significant positive association between UNC13B levels and lysosome formation in the 17.94 cell line. Lysosomes likely serve a role in the sensitivity of Wilms' tumor cell lines to drugs. Elevated UNC13B expression was observed in the 17.94 Wilms' tumor cell line compared to normal kidney cells. UNC13B knockdown also resulted in increased apoptosis levels upon doxorubicin treatment. Immunofluorescence revealed UNC13B localization within cellular vesicles, and its knockdown significantly decreased lysosome levels. Overall, the findings of the present study demonstrate that UNC13B regulates the sensitivity of the Wilms' tumor 17.94 cell line to doxorubicin by modulating lysosome formation within cells. The results suggest that UNC13B is likely an enriched target involved in lysosomal regulation in certain tumors, offering a new approach for optimizing chemotherapy in Wilms' tumor and other cancers with high UNC13B expression.
Collapse
Affiliation(s)
- Xi Chen
- Department of Neonatology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Yingying Bao
- Department of Neonatology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Ge Sun
- Department of Neonatology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Xiaobo Wang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong 518107, P.R. China
| | - Jiajun Zhu
- Department of Neonatology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
- Zhejiang Provincial Clinical Research Center for Child Health, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
4
|
Gong F, Cui Y, Lv P, Liu J, Sun X, Han P, Zhou L, Xia T, Cao W. Role of ESCCAL-1 in regulating exocytosis of AuNPs in human esophageal squamous carcinoma cells. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 59:102754. [PMID: 38797223 DOI: 10.1016/j.nano.2024.102754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 04/19/2024] [Accepted: 05/06/2024] [Indexed: 05/29/2024]
Abstract
Exocytosis is a critical factor for designing efficient nanocarriers and determining cytotoxicity. However, the research on the exocytosis mechanism of nanoparticles, especially the role of long non-coding RNAs (lncRNAs), has not been reported. In this study, the exocytosis of AuNPs in the KYSE70 cells and the involved molecular pathways of exocytosis are analyzed. It demonstrates that nanoparticles underwent time-dependent release from the cells by exocytosis, and the release of β-hexosaminidase confirms that AuNPs are excreted through lysosomes. Mechanistic studies reveal that lncRNA ESCCAL-1 plays a vital role in controlling the exocytosis of AuNPs through activation of the MAPK pathway, including the phosphorylation of ERK and JNK. The study implies that the ESCCAL-1-mediated pathway plays an important role in the exocytosis of AuNPs in KYSE70 cells. This finding has implications for the role of ESCCAL-1 on the drug resistance of esophagus cancer by controlling lysosome-mediated exocytosis.
Collapse
Affiliation(s)
- Fenfen Gong
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Yuanbo Cui
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Pengju Lv
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Jia Liu
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Xiaoyan Sun
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Pengli Han
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Lijuan Zhou
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Tian Xia
- Division of NanoMedicine, Department of Medicine, University of California, Los Angeles, 90095 Los Angeles, CA, United States.
| | - Wei Cao
- Translational Medical Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China; Henan Province Joint International Laboratory for Bioconjugation and Antibody Coupling, Zhengzhou, Henan 450001, China.
| |
Collapse
|
5
|
Chang X, Wang WX. Differential cellular uptake and trafficking of nanoplastics in two hemocyte subpopulations of mussels Perna viridis. JOURNAL OF HAZARDOUS MATERIALS 2024; 471:134388. [PMID: 38669925 DOI: 10.1016/j.jhazmat.2024.134388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/27/2024] [Accepted: 04/21/2024] [Indexed: 04/28/2024]
Affiliation(s)
- Xinyi Chang
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China; Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
| | - Wen-Xiong Wang
- School of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China; Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China.
| |
Collapse
|
6
|
Yasamineh S, Mehrabani FJ, Derafsh E, Danihiel Cosimi R, Forood AMK, Soltani S, Hadi M, Gholizadeh O. Potential Use of the Cholesterol Transfer Inhibitor U18666A as a Potent Research Tool for the Study of Cholesterol Mechanisms in Neurodegenerative Disorders. Mol Neurobiol 2024; 61:3503-3527. [PMID: 37995080 DOI: 10.1007/s12035-023-03798-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 11/03/2023] [Indexed: 11/24/2023]
Abstract
Cholesterol is an essential component of mammalian cell membranes and a precursor for crucial signaling molecules. The brain contains the highest level of cholesterol in the body, and abnormal cholesterol metabolism links to many neurodegenerative disorders. The results indicate that faulty cholesterol metabolism is a common feature among people living with neurodegenerative conditions. The researchers suggest that restoring cholesterol levels may become a beneficial new strategy in treating certain neurodegenerative conditions. Several neurodegenerative disorders, such as Alzheimer's disease (AD), Niemann-Pick type C (NPC) disease, and Parkinson's disease (PD), have been connected to abnormalities in brain cholesterol metabolism. Consequently, using a lipid research tool is vital to study further and understand the effect of lipids in neurodegenerative disorders such as NPC, AD, PD, and Huntington's disease (HD). U18666A, also known as 3-(2-(diethylamino) ethoxy) androst-5-en-17-one, is a pharmaceutical drug that suppresses cholesterol trafficking and is a well-known class-2 amphiphile. U18666A has performed many functions, allowing for essential discoveries in lipid studies and shedding light on the pathophysiology of neurodegenerative disorders. Additionally, U18666A prevented the downregulation of low-density lipoprotein (LDL) receptors that are induced by LDL and led to the buildup of cholesterol in lysosomes. Numerous studies show that U18666A impacts the function of cholesterol trafficking to control the metabolism and transport of amyloid precursor proteins (APPs). Treating cortical neurons with U18666A may provide a new in vitro model system for studying the underlying molecular process of NPC, AD, HD, and PD. In this article, we review the mechanism and function of U18666A as a vital tool for studying cholesterol mechanisms in neurological diseases related to abnormal cholesterol metabolism, such as AD, NPC, HD, and PD.
Collapse
Affiliation(s)
| | | | - Ehsan Derafsh
- Windsor University School of Medicine, Cayon, Saint Kitts and Nevis
| | | | | | - Siamak Soltani
- Department of Forensic Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Meead Hadi
- Department Of Microbiology, Faculty of Basic Sciences, Tehran Central Branch, Islamic Azad University, Tehran, Iran
| | | |
Collapse
|
7
|
Huang Z, Zhou Z, Ye Q, Li X, Wang T, Li J, Dong W, Guo R, Ding Y, Xue H, Ding H, Lau CH. Effects of Different Surface Functionalizations of Silica Nanoparticles on Mesenchymal Stem Cells. ACS APPLIED BIO MATERIALS 2024; 7:3295-3305. [PMID: 38701399 DOI: 10.1021/acsabm.4c00241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Physicochemical properties of nanoparticles, such as particle size, surface charge, and particle shape, have a significant impact on cell activities. However, the effects of surface functionalization of nanoparticles with small chemical groups on stem cell behavior and function remain understudied. Herein, we incorporated different chemical functional groups (amino, DETA, hydroxyl, phosphate, and sulfonate with charges of +9.5, + 21.7, -14.1, -25.6, and -37.7, respectively) to the surface of inorganic silica nanoparticles. To trace their effects on mesenchymal stem cells (MSCs) of rat bone marrow, these functionalized silica nanoparticles were used to encapsulate Rhodamine B fluorophore dye. We found that surface functionalization with positively charged and short-chain chemical groups facilitates cell internalization and retention of nanoparticles in MSCs. The endocytic pathway differed among functionalized nanoparticles when tested with ion-channel inhibitors. Negatively charged nanoparticles mainly use lysosomal exocytosis to exit cells, while positively charged nanoparticles can undergo endosomal escape to avoid scavenging. The cytotoxic profiles of these functionalized silica nanoparticles are still within acceptable limits and tolerable. They exerted subtle effects on the actin cytoskeleton and migration ability. Last, phosphate-functionalized nanoparticles upregulate osteogenesis-related genes and induce osteoblast-like morphology, implying that it can direct MSCs lineage specification for bone tissue engineering. Our study provides insights into the rational design of biomaterials for effective drug delivery and regenerative medicine.
Collapse
Affiliation(s)
- Zhihao Huang
- Department of Biology, College of Science, Shantou University, 515063 Shantou, Guangdong, China
| | - Zhongqi Zhou
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107 Shenzhen, Guangdong, China
| | - Qiaoyuan Ye
- Department of Dermatology, The Second Clinical Medical College, Guangdong Medical University, 523808 Dongguan, Guangdong, China
| | - Xiaoyan Li
- Center for Vascular Surgery and Wound Care, Jinshan Hospital, Fudan University, 200540 Shanghai, China
| | - Tao Wang
- Department of Biology, College of Science, Shantou University, 515063 Shantou, Guangdong, China
| | - Jiaqi Li
- Department of Biology, College of Science, Shantou University, 515063 Shantou, Guangdong, China
| | - Wenjiao Dong
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Medical University, 523808 Dongguan, Guangdong, China
| | - Rui Guo
- Animal Husbandry and Veterinary Institute, Hubei Academy of Agricultural Science, 430064 Wuhan, Hubei, China
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis, Ministry of Agriculture, 430064 Wuhan, Hubei, China
| | - Yuanlin Ding
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Medical University, 523808 Dongguan, Guangdong, China
| | - Hongman Xue
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107 Shenzhen, Guangdong, China
| | - Haifeng Ding
- Department of Otolaryngology, Shenzhen Pingshan District People's Hospital, 518118 Shenzhen, Guangdong, China
| | - Cia-Hin Lau
- Department of Biology, College of Science, Shantou University, 515063 Shantou, Guangdong, China
| |
Collapse
|
8
|
Lu YY, Lu L, Ren HY, Hua W, Zheng N, Huang FY, Wang J, Tian M, Huang Q. The size-dependence and reversibility of polystyrene nanoplastics-induced lipid accumulation in mice: Possible roles of lysosomes. ENVIRONMENT INTERNATIONAL 2024; 185:108532. [PMID: 38422876 DOI: 10.1016/j.envint.2024.108532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/02/2024] [Accepted: 02/22/2024] [Indexed: 03/02/2024]
Abstract
Nanoplastics (NPs) continue to accumulate in global aquatic and terrestrial systems, posing a potential threat to human health through the food chain and/or other pathways. Both in vivo and in vitro studies have confirmed that the liver is one of the main organs targeted for the accumulation of NPs in living organisms. However, whether exposure to NPs induces size-dependent disorders of liver lipid metabolism remains controversial, and the reversibility of NPs-induced hepatotoxicity is largely unknown. In this study, the effects of long-term exposure to environmentally relevant doses of polystyrene nanoplastics (PS-NPs) on lipid accumulation were investigated in terms of autophagy and lysosomal mechanisms. The findings indicated that hepatic lipid accumulation was more pronounced in mice exposed to 100 nm PS-NPs compared to 500 nm PS-NPs. This effect was effectively alleviated after 50 days of self-recovery for 100 nm and 500 nm PS-NPs exposure. Mechanistically, although PS-NPs exposure activated autophagosome formation through ERK (mitogen-activated protein kinase 1)/mTOR (mechanistic target of rapamycin kinase) signaling pathway, the inhibition of Rab7 (RAB7, member RAS oncogene family), CTSB (cathepsin B), and CTSD (cathepsin D) expression impaired lysosomal function, thereby blocking autophagic flux and contributing to hepatic lipid accumulation. After termination of PS-NPs exposure, lysosomal exocytosis was responsible for the clearance of PS-NPs accumulated in lysosomes. Furthermore, impaired lysosomal function and autophagic flux inhibition were effectively alleviated. This might be the main reason for the alleviation of PS-NPs-induced lipid accumulation after recovery. Collectively, we demonstrate for the first time that lysosomes play a dual role in the persistence and reversibility of hepatotoxicity induced by environmental relevant doses of NPs, which provide novel evidence for the prevention and intervention of liver injury associated with nanoplastics exposure.
Collapse
Affiliation(s)
- Yan-Yang Lu
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China
| | - Lu Lu
- College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Hong-Yun Ren
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China
| | - Weizhen Hua
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Nengxing Zheng
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Fu-Yi Huang
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China
| | - Jiani Wang
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China
| | - Meiping Tian
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China
| | - Qingyu Huang
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, China.
| |
Collapse
|
9
|
Peng Y, Yang Z, Sun H, Li J, Lan X, Liu S. Nanomaterials in Medicine: Understanding Cellular Uptake, Localization, and Retention for Enhanced Disease Diagnosis and Therapy. Aging Dis 2024:AD.2024.0206-1. [PMID: 38421835 DOI: 10.14336/ad.2024.0206-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
Nanomaterials (NMs) have emerged as promising tools for disease diagnosis and therapy due to their unique physicochemical properties. To maximize the effectiveness and design of NMs-based medical applications, it is essential to comprehend the complex mechanisms of cellular uptake, subcellular localization, and cellular retention. This review illuminates the various pathways that NMs take to get from the extracellular environment to certain intracellular compartments by investigating the various mechanisms that underlie their interaction with cells. The cellular uptake of NMs involves complex interactions with cell membranes, encompassing endocytosis, phagocytosis, and other active transport mechanisms. Unique uptake patterns across cell types highlight the necessity for customized NMs designs. After internalization, NMs move through a variety of intracellular routes that affect where they are located subcellularly. Understanding these pathways is pivotal for enhancing the targeted delivery of therapeutic agents and imaging probes. Furthermore, the cellular retention of NMs plays a critical role in sustained therapeutic efficacy and long-term imaging capabilities. Factors influencing cellular retention include nanoparticle size, surface chemistry, and the cellular microenvironment. Strategies for prolonging cellular retention are discussed, including surface modifications and encapsulation techniques. In conclusion, a comprehensive understanding of the mechanisms governing cellular uptake, subcellular localization, and cellular retention of NMs is essential for advancing their application in disease diagnosis and therapy. This review provides insights into the intricate interplay between NMs and biological systems, offering a foundation for the rational design of next-generation nanomedicines.
Collapse
Affiliation(s)
- Yue Peng
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Zhengshuang Yang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Hui Sun
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Jinling Li
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiuwan Lan
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Sijia Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine & Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research & Guangxi Key Laboratory of Brain Science, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
10
|
Guo J, Xu S, Majeed U, Ye J, Zhang H, Xue W, Luo Y. Size-Related Pathway Flux Analysis of Ultrasmall Iron Oxide Nanoparticles in Macrophage Cell RAW264.7 for Safety Evaluation. ACS OMEGA 2024; 9:3480-3490. [PMID: 38284085 PMCID: PMC10809237 DOI: 10.1021/acsomega.3c07081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/20/2023] [Accepted: 12/25/2023] [Indexed: 01/30/2024]
Abstract
The endocytosis, intracellular transport, and exocytosis of different-sized nanoparticles were reported to greatly affect their efficacy and biosafety. The quantitation of endocytosis and exocytosis as well as subcellular distribution of nanoparticles might be an effective approach based on transport pathway flux analysis. Thus, the key parameters that could present the effects of three different-sized ultrasmall iron oxide nanoparticles (USIONPs) were systematically investigated in RAW264.7 cells. The endocytosis and exocytosis of USIONPs were related to their sizes; 15.4 nm of S2 could be quickly and more internalized and excreted in comparison to S1 (7.8 nm) and S3 (30.7 nm). In RAW264.7 cells, USIONPs were observed in endosomes, lysosomes, the Golgi apparatus, and autophagosomes via a transmission electron microscope. Based on flux analysis of intracellular transport pathways of USIONPs, it was found that 43% of S1, 40% of S2, and 44% of S3 were individually transported extracellularly through the Golgi apparatus-involved middle-fast pathway, while 24% of S1, 23% of S2, and 26% of S3 were transported through the fast recycling endosomal pathway, and the residues were transported through the slower speed lysosomal pathway. USIONPs might be transported via size-related endocytosis and exocytosis pathways. The pathway flux could be calculated on the basis of disturbance analysis of special transporters as well as their coding genes. Because there were rate differences among these transport pathways, this pathway flux could anticipate the intracellular remaining time and distribution of different-sized nanoparticles, the function exertion, and side effects of nanomaterials. The size of the nanomaterials could be optimized for improving functions and safety.
Collapse
Affiliation(s)
- Jiaqing Guo
- School of Chemical Engineering, Northwest University, Xi'an 710069, China
| | - Shixin Xu
- School of Chemical Engineering, Northwest University, Xi'an 710069, China
| | - Usman Majeed
- College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Jianming Ye
- College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Huaxin Zhang
- School of Chemical Engineering, Northwest University, Xi'an 710069, China
| | - Weiming Xue
- School of Chemical Engineering, Northwest University, Xi'an 710069, China
| | - Yane Luo
- College of Food Science and Technology, Northwest University, Xi'an 710069, China
| |
Collapse
|
11
|
Zhang Y, Jia Z, Gao X, Zhao J, Zhang H. Polystyrene nanoparticles induced mammalian intestine damage caused by blockage of BNIP3/NIX-mediated mitophagy and gut microbiota alteration. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 907:168064. [PMID: 37884137 DOI: 10.1016/j.scitotenv.2023.168064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 10/01/2023] [Accepted: 10/21/2023] [Indexed: 10/28/2023]
Abstract
Nanoplastics possess the capacity for cellular internalization, and consequentially disrupt mitochondrial functionality, precipitating aberrations in energy metabolism. Given this, the potential accumulation of nanoplastics in alimentary sources presents a considerable hazard to the mammalian gastrointestinal system. While mitophagy serves as a cytoprotective mechanism that sustains redox homeostasis through the targeted removal of compromised mitochondria, the regulatory implications of mitophagy in nanoplastic-induced toxicity remain an underexplored domain. In the present investigation, polystyrene (PS) nanoparticles, with a diameter of 80 nm employed as a representative model to assess their toxicological impact and propensity to instigate mitophagy in intestinal cells both in vitro and in vivo. Data indicated that PS nanoparticles elicited BNIP3/NIX-mediated mitophagy within the intestinal milieu. Strikingly, the impediment of this degradation process at elevated concentrations was correlated with exacerbated pathological ramifications. In vitro assays corroborated that high-dosage cellular uptake of PS nanoparticles obstructed the mitophagy pathway. Furthermore, treatment with PS nanoparticles engendered alterations in gut microbiota composition and manifested a proclivity to modulate nutritional metabolism. Collectively, these findings elucidate that oral exposure to PS nanoparticles culminates in the inhibition of mitophagy and induces perturbations in the intestinal microbiota. This contributes valuable insights into the toxicological repercussions of nanoplastics on mammalian gastrointestinal health.
Collapse
Affiliation(s)
- Yilun Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Science, Shandong Normal University, Jinan, Shandong 250014, China
| | - Zhenzhen Jia
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Science, Shandong Normal University, Jinan, Shandong 250014, China
| | - Xianlei Gao
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Juan Zhao
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Science, Shandong Normal University, Jinan, Shandong 250014, China
| | - Hongyan Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Science, Shandong Normal University, Jinan, Shandong 250014, China.
| |
Collapse
|
12
|
Shirbhate E, Singh V, Mishra A, Jahoriya V, Veerasamy R, Tiwari AK, Rajak H. Targeting Lysosomes: A Strategy Against Chemoresistance in Cancer. Mini Rev Med Chem 2024; 24:1449-1468. [PMID: 38343053 DOI: 10.2174/0113895575287242240129120002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/11/2024] [Accepted: 01/19/2024] [Indexed: 07/23/2024]
Abstract
Chemotherapy is still the major method of treatment for many types of cancer. Curative cancer therapy is hampered significantly by medication resistance. Acidic organelles like lysosomes serve as protagonists in cellular digestion. Lysosomes, however, are gaining popularity due to their speeding involvement in cancer progression and resistance. For instance, weak chemotherapeutic drugs of basic nature permeate through the lysosomal membrane and are retained in lysosomes in their cationic state, while extracellular release of lysosomal enzymes induces cancer, cytosolic escape of lysosomal hydrolases causes apoptosis, and so on. Drug availability at the sites of action is decreased due to lysosomal drug sequestration, which also enhances cancer resistance. This review looks at lysosomal drug sequestration mechanisms and how they affect cancer treatment resistance. Using lysosomes as subcellular targets to combat drug resistance and reverse drug sequestration is another method for overcoming drug resistance that is covered in this article. The present review has identified lysosomal drug sequestration as one of the reasons behind chemoresistance. The article delves deeper into specific aspects of lysosomal sequestration, providing nuanced insights, critical evaluations, or novel interpretations of different approaches that target lysosomes to defect cancer.
Collapse
Affiliation(s)
- Ekta Shirbhate
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Vaibhav Singh
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Aditya Mishra
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Varsha Jahoriya
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Ravichandran Veerasamy
- Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Amit K Tiwari
- UAMS College of Pharmacy; UAMS - University of Arkansas for Medical Sciences, (AR) USA
| | - Harish Rajak
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| |
Collapse
|
13
|
da Costa Marques R, Hüppe N, Speth KR, Oberländer J, Lieberwirth I, Landfester K, Mailänder V. Proteomics reveals time-dependent protein corona changes in the intracellular pathway. Acta Biomater 2023; 172:355-368. [PMID: 37839632 DOI: 10.1016/j.actbio.2023.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/17/2023]
Abstract
The intracellular protein corona has not been fully investigated in the field of nanotechnology-biology (nano-bio) interactions. To effectively understand intracellular protein corona formation and dynamics, we established a workflow to isolate the intracellular protein corona at different uptake times of two nanoparticles - magnetic hydroxyethyl starch nanoparticles (HES-NPs) and magnetic human serum albumin nanocapsules (HSA-NCs). We performed label-free quantitative LC-MS proteomics to analyze the composition of the intracellular protein corona and correlated our findings with results from conventional methods for intracellular trafficking of nanocarriers, such as flow cytometry, transmission electron microscopy (TEM), and confocal microscopy (cLSM). We determined the evolution of the intracellular protein corona. At different time stages the protein corona of the HES-NPs with a slower uptake changed, but there were fewer changes in that of the HSA-NCs with a more rapid uptake. We identified proteins that are involved in macropinocytosis (RAC1, ASAP2) as well as caveolin. This was confirmed by blocking experiments and by TEM studies. The investigated nanocarrier predominantly trafficked from early endosomes as determined by RAB5 identification in proteomics and in cLSM to late endosomes/lysosomes (RAB7, LAMP1, cathepsin K and HSP 90-beta) We further demonstrated differences between nanoparticles with slower and faster uptake kinetics and determined the associated proteome at different time points. Analysis of the intracellular protein corona provides us with effective data to examine the intracellular trafficking of nanocarriers used in efficient drug delivery and intracellular applications. STATEMENT OF SIGNIFICANCE: Many research papers focus on the protein corona on nanoparticles formed in biological fluids, but there are hardly any articles dealing with proteins that come in contact with nanoparticles inside cells. The "intracellular protein corona" studied here is a far more complex and highly demanding field. Most nanocarriers are designed to be taken up into cells. Given this, we chose two different nanocarriers to reveal changes in the proteins in dendritic cells during contact at specific times. Further studies will allow us to examine molecular target proteins using these methods. Our research is a significant addition towards the goal of understanding and thus improving the efficacy of drug nanocarriers.
Collapse
Affiliation(s)
- Richard da Costa Marques
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany; Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Natkritta Hüppe
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Kai R Speth
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany; Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Jennifer Oberländer
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany; Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Ingo Lieberwirth
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Katharina Landfester
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Volker Mailänder
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany; Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany.
| |
Collapse
|
14
|
Singh N, Mudassir M, Ansari S, Chosdol K, Sinha S, Chattopadhyay P. Poly(lactic-co-glycolic) acid nanoparticles localize in vesicles after diffusing into cells and are retained by intracellular traffic modulators. Nanomedicine (Lond) 2023; 18:1907-1919. [PMID: 38078434 DOI: 10.2217/nnm-2023-0139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023] Open
Abstract
Aim: We investigated our previous finding of increased retention of poly(lactic-co-glycolic) acid nanoparticles (PLGA-NPs) with metabolic inhibitors (MI) and studied the effect of some small molecule inhibitors on PLGA-NP assimilation. Materials & methods: Intracellular PLGA-NP colocalization in the presence of MI was investigated by confocal microscopy. Intracellular retention of PLGA-NPs by some small molecules was estimated by fluorescence microscopy and flow cytometry after Pulse/Chase experiments. Results: MI caused PLGA-NP colocalization in intracellular membranous structures, mainly endosomes and lysosomes. Some small molecule inhibitors demonstrated increased intracellular PLGA-NP accumulation. Conclusion: This study elucidates the movement of PLGA-NP in cells and suggests that clinically used small molecules can reduce their extrusion by enhancing their stay within intracellular vesicles, with possible clinically beneficial consequences.
Collapse
Affiliation(s)
- Neha Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Madeeha Mudassir
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
- Department of Obstetrics and Gynaecology, University College of Medical Sciences, GTB Hospital, Delhi, 110095, India
| | - Shiba Ansari
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
- Department of Biochemistry, University College of Medical Sciences, GTB Hospital, Delhi, 110095, India
| | - Kunzang Chosdol
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Subrata Sinha
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | | |
Collapse
|
15
|
Onodera A, Shimomura T, Ochi H, Sunada R, Fukutomi E, Hidaka K, Kawai Y. The Cellular Accumulation of Vehicle Exhaust Particulates Changes the Acidic pH Environment of Lysosomes in BEAS-2B Airway Epithelial Cells. J Xenobiot 2023; 13:653-661. [PMID: 37987443 PMCID: PMC10660702 DOI: 10.3390/jox13040042] [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: 09/25/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/22/2023] Open
Abstract
Many people are exposed every day to vehicle exhaust particulates (VEPs), which are thought to be taken up by epithelial cells that are the first barrier in our biological defense. The study aim was to investigate how VEPs are processed in the lysosomal degradation system. BEAS-2B airway epithelial cells easily ingest VEPs and have been shown to accumulate in cells for several days, but no elevated cytotoxicity was observed over that time period. An analysis of 3D images confirmed the presence of VEPs in or near lysosomes, and an accumulation of VEPs resulted in an increase in the normal acidic pH in lysosomes and the extracellular release of the lysosomal enzyme β-hexosaminidase. Epithelial cells were thought to activate the lysosome-mediated secretion of extracellular vesicles to avoid damage caused by non-degradable foreign substances, such as VEPs, and as a side reaction, the acidic pH environment of the lysosomes could not be maintained.
Collapse
Affiliation(s)
- Akira Onodera
- Department of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe 650-8586, Japan; (T.S.); (H.O.); (R.S.); (E.F.); (Y.K.)
| | - Takuya Shimomura
- Department of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe 650-8586, Japan; (T.S.); (H.O.); (R.S.); (E.F.); (Y.K.)
| | - Hirohisa Ochi
- Department of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe 650-8586, Japan; (T.S.); (H.O.); (R.S.); (E.F.); (Y.K.)
| | - Ryuto Sunada
- Department of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe 650-8586, Japan; (T.S.); (H.O.); (R.S.); (E.F.); (Y.K.)
| | - Eiko Fukutomi
- Department of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe 650-8586, Japan; (T.S.); (H.O.); (R.S.); (E.F.); (Y.K.)
| | - Koushi Hidaka
- Research Facility Center for Science and Technology, Kobe University, 1-1, Rokkodai-cho, Nada-ku, Kobe 657-8501, Japan;
| | - Yuichi Kawai
- Department of Pharmaceutical Sciences, Kobe Gakuin University, 1-1-3 Minatojima, Chuo-ku, Kobe 650-8586, Japan; (T.S.); (H.O.); (R.S.); (E.F.); (Y.K.)
| |
Collapse
|
16
|
Shang L, Xie Q, Yang C, Kong L, Zhang Z. Extracellular Vesicles Facilitate the Transportation of Nanoparticles within and between Cells for Enhanced Tumor Therapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:42378-42394. [PMID: 37658814 DOI: 10.1021/acsami.3c10237] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
The interaction between nanoparticles and cells is closely associated with the therapeutic effects of nanomedicine. Nanoparticles could be transported among cells, but the process-related mechanism remains to be further explored. In this study, it was found that endocytosed cationic polymer nanoparticles (cNPs) could be excreted in an extracellular vesicle (EV)-coated form (cNP@EVs). It was deduced that cNPs may pass through early endosomes, multivesicular bodies (MVBs), and autophagic MVBs within cells. Moreover, a high level of autophagy facilitated the exocytosis process. Since EVs were the effective vehicles for conveying biological information and substances, cNP@EVs were proved to be efficient forms for the intercellular transportation of nanoparticles and have the potential as efficient biomimetic drug delivery systems. These properties endowed cNP@EVs with deep penetration and enhanced antitumor activity. Our findings provided a proof-of-concept for understanding the transfer process of nanoparticles among cells and may help us to further utilize EV-mediated transportation of nanoparticles, therefore, expanding its clinical application.
Collapse
Affiliation(s)
- Lihuan Shang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Xie
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Conglian Yang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Kong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Engineering Research Centre for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
17
|
Liu J, Liu YY, Li CS, Cao A, Wang H. Exocytosis of Nanoparticles: A Comprehensive Review. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2215. [PMID: 37570533 PMCID: PMC10421347 DOI: 10.3390/nano13152215] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023]
Abstract
Both biomedical applications and safety assessments of manufactured nanomaterials require a thorough understanding of the interaction between nanomaterials and cells, including how nanomaterials enter cells, transport within cells, and leave cells. However, compared to the extensively studied uptake and trafficking of nanoparticles (NPs) in cells, less attention has been paid to the exocytosis of NPs. Yet exocytosis is an indispensable process of regulating the content of NPs in cells, which in turn influences, even decides, the toxicity of NPs to cells. A comprehensive understanding of the mechanisms and influencing factors of the exocytosis of NPs is not only essential for the safety assessment of NPs but also helpful for guiding the design of safe and highly effective NP-based materials for various purposes. Herein, we review the current status and progress of studies on the exocytosis of NPs. Firstly, we introduce experimental procedures and considerations. Then, exocytosis mechanisms/pathways are summarized with a detailed introduction of the main pathways (lysosomal and endoplasmic reticulum/Golgi pathway) and the role of microtubules; the patterns of exocytosis kinetics are presented and discussed. Subsequently, the influencing factors (initial content and location of intracellular NPs, physiochemical properties of NPs, cell type, and extracellular conditions) are fully discussed. Although there are inconsistent results, some rules are obtained, like smaller and charged NPs are more easily excreted. Finally, the challenges and future directions in the field have been discussed.
Collapse
Affiliation(s)
| | | | | | | | - Haifang Wang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| |
Collapse
|
18
|
Abstract
Nanoparticles (NPs) have been widely used in different areas, including consumer products and medicine. In terms of biomedical applications, NPs or NP-based drug formulations have been extensively investigated for cancer diagnostics and therapy in preclinical studies, but the clinical translation rate is low. Therefore, a thorough and comprehensive understanding of the pharmacokinetics of NPs, especially in drug delivery efficiency to the target therapeutic tissue tumor, is important to design more effective nanomedicines and for proper assessment of the safety and risk of NPs. This review article focuses on the pharmacokinetics of both organic and inorganic NPs and their tumor delivery efficiencies, as well as the associated mechanisms involved. We discuss the absorption, distribution, metabolism, and excretion (ADME) processes following different routes of exposure and the mechanisms involved. Many physicochemical properties and experimental factors, including particle type, size, surface charge, zeta potential, surface coating, protein binding, dose, exposure route, species, cancer type, and tumor size can affect NP pharmacokinetics and tumor delivery efficiency. NPs can be absorbed with varying degrees following different exposure routes and mainly accumulate in liver and spleen, but also distribute to other tissues such as heart, lung, kidney and tumor tissues; and subsequently get metabolized and/or excreted mainly through hepatobiliary and renal elimination. Passive and active targeting strategies are the two major mechanisms of tumor delivery, while active targeting tends to have less toxicity and higher delivery efficiency through direct interaction between ligands and receptors. We also discuss challenges and perspectives remaining in the field of pharmacokinetics and tumor delivery efficiency of NPs.
Collapse
Affiliation(s)
- Long Yuan
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32610, USA
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32608, USA
| | - Qiran Chen
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32610, USA
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32608, USA
| | - Jim E. Riviere
- 1Data Consortium, Kansas State University, Olathe, KS 66061, USA
| | - Zhoumeng Lin
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32610, USA
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32608, USA
| |
Collapse
|
19
|
Singh N, Kim J, Kim J, Lee K, Zunbul Z, Lee I, Kim E, Chi SG, Kim JS. Covalent organic framework nanomedicines: Biocompatibility for advanced nanocarriers and cancer theranostics applications. Bioact Mater 2023; 21:358-380. [PMID: 36185736 PMCID: PMC9483748 DOI: 10.1016/j.bioactmat.2022.08.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 11/19/2022] Open
Abstract
Nanomedicines for drug delivery and imaging-guided cancer therapy is a rapidly growing research area. The unique properties of nanomedicines have a massive potential in solving longstanding challenges of existing cancer drugs, such as poor localization at the tumor site, high drug doses and toxicity, recurrence, and poor immune response. However, inadequate biocompatibility restricts their potential in clinical translation. Therefore, advanced nanomaterials with high biocompatibility and enhanced therapeutic efficiency are highly desired to fast-track the clinical translation of nanomedicines. Intrinsic properties of nanoscale covalent organic frameworks (nCOFs), such as suitable size, modular pore geometry and porosity, and straightforward post-synthetic modification via simple organic transformations, make them incredibly attractive for future nanomedicines. The ability of COFs to disintegrate in a slightly acidic tumor microenvironment also gives them a competitive advantage in targeted delivery. This review summarizes recently published applications of COFs in drug delivery, photo-immuno therapy, sonodynamic therapy, photothermal therapy, chemotherapy, pyroptosis, and combination therapy. Herein we mainly focused on modifications of COFs to enhance their biocompatibility, efficacy and potential clinical translation. This review will provide the fundamental knowledge in designing biocompatible nCOFs-based nanomedicines and will help in the rapid development of cancer drug carriers and theranostics.
Collapse
Affiliation(s)
- Nem Singh
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Jungryun Kim
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Jaewon Kim
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Kyungwoo Lee
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Zehra Zunbul
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Injun Lee
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Eunji Kim
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| | - Sung-Gil Chi
- Department of Life Science, Korea University, Seoul, 02841, South Korea
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul, 02841, South Korea
| |
Collapse
|
20
|
Mlejnek P. What Is the Significance of Lysosomal-Mediated Resistance to Imatinib? Cells 2023; 12:cells12050709. [PMID: 36899844 PMCID: PMC10000661 DOI: 10.3390/cells12050709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
The lysosomal sequestration of hydrophobic weak-base anticancer drugs is one proposed mechanism for the reduced availability of these drugs at target sites, resulting in a marked decrease in cytotoxicity and consequent resistance. While this subject is receiving increasing emphasis, it is so far only in laboratory experiments. Imatinib is a targeted anticancer drug used to treat chronic myeloid leukaemia (CML), gastrointestinal stromal tumours (GISTs), and a number of other malignancies. Its physicochemical properties make it a typical hydrophobic weak-base drug that accumulates in the lysosomes of tumour cells. Further laboratory studies suggest that this might significantly reduce its antitumor efficacy. However, a detailed analysis of published laboratory studies shows that lysosomal accumulation cannot be considered a clearly proven mechanism of resistance to imatinib. Second, more than 20 years of clinical experience with imatinib has revealed a number of resistance mechanisms, none of which is related to its accumulation in lysosomes. This review focuses on the analysis of salient evidence and raises a fundamental question about the significance of lysosomal sequestration of weak-base drugs in general as a possible resistance mechanism both in clinical and laboratory settings.
Collapse
Affiliation(s)
- Petr Mlejnek
- Department of Anatomy, Faculty of Medicine and Dentistry, Palacky University Olomouc, Hnevotinska 3, 77515 Olomouc, Czech Republic
| |
Collapse
|
21
|
Bonamy C, Pesnel S, Ben Haddada M, Gorgette O, Schmitt C, Morel AL, Sauvonnet N. Impact of Green Gold Nanoparticle Coating on Internalization, Trafficking, and Efficiency for Photothermal Therapy of Skin Cancer. ACS OMEGA 2023; 8:4092-4105. [PMID: 36743010 PMCID: PMC9893490 DOI: 10.1021/acsomega.2c07054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/29/2022] [Indexed: 06/18/2023]
Abstract
Skin cancer is a global health issue and mainly composed of melanoma and nonmelanoma cancers. For the first clinical proof of concept on humans, we decided to study good prognosis skin cancers, i.e., carcinoma basal cell. In UE, the first-line treatment remains surgical resection, healing most of the tumors, but presents aesthetic disadvantages with a high reoccurrence rate on exposed areas. Moreover, the therapeutic indications could extend to melanoma and metastasis, which is a different medical strategy that could combine this treatment. Indeed, patients with late-stage melanoma are in a therapeutic impasse, despite recent targeted and immunological therapies. Photothermal therapy using gold nanoparticles is the subject of many investigations due to their strong potential to treat cancers by physical, thermal destruction. We developed gold nanoparticles synthesized by green chemistry (gGNPs), using endemic plant extract from Reunion Island, which have previously showed their efficiency at a preclinical stage. Here, we demonstrate that these gGNPs are less cytotoxic than gold nanoparticles synthesized by Turkevich's method. Furthermore, our work describes the optimization of gGNP coating and stabilization, also taking into consideration the gGNP path in cells (endocytosis, intracellular trafficking, and exocytosis), their specificity toward cancerous cells, their cytotoxicity, and their in vivo efficiency. Finally, based on the metabolic switch of cancerous cells overexpressing Glut transporters in skin cancers, we demonstrated that glucose-stabilized gGNP (gGNP@G) enables a quick internalization, fourfold higher in cancerous cells in contrast to healthy cells with no side cytotoxicity, which is particularly relevant to target and treat cancer.
Collapse
Affiliation(s)
- Clément Bonamy
- Torskal, 2 rue Maxime Rivière, 97490 Sainte-Clotilde, France
- Group
Intracellular Trafficking and Tissue Homeostasis, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Sabrina Pesnel
- Torskal, 2 rue Maxime Rivière, 97490 Sainte-Clotilde, France
| | | | - Olivier Gorgette
- Ultrastructural
BioImaging, Institut Pasteur, Université
Paris Cité, 75015 Paris, France
| | - Christine Schmitt
- Ultrastructural
BioImaging, Institut Pasteur, Université
Paris Cité, 75015 Paris, France
| | | | - Nathalie Sauvonnet
- Group
Intracellular Trafficking and Tissue Homeostasis, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| |
Collapse
|
22
|
Luo Z, Wu S, Zhou J, Xu W, Xu Q, Lu L, Xie C, Liu Y, Lu W. All-stage targeted therapy for the brain metastasis from triple-negative breast cancer. Acta Pharm Sin B 2023; 13:359-371. [PMID: 36815053 PMCID: PMC9939358 DOI: 10.1016/j.apsb.2022.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 11/27/2022] Open
Abstract
Brain metastasis is a common and serious complication of breast cancer, which is commonly associated with poor survival and prognosis. In particular, the treatment of brain metastasis from triple-negative breast cancer (BM-TNBC) has to face the distinct therapeutic challenges from tumor heterogeneity, circulating tumor cells (CTCs), blood-brain barrier (BBB) and blood-tumor barrier (BTB), which is in unmet clinical needs. Herein, combining with the advantages of synthetic and natural targeting moieties, we develop a "Y-shaped" peptide pVAP-decorated platelet-hybrid liposome drug delivery system to address the all-stage targeted drug delivery for the whole progression of BM-TNBC. Inherited from the activated platelet, the hybrid liposomes still retain the native affinity toward CTCs. Further, the peptide-mediated targeting to breast cancer cells and transport across BBB/BTB are demonstrated in vitro and in vivo. The resultant delivery platform significantly improves the drug accumulation both in orthotopic breast tumors and brain metastatic lesions, and eventually exhibits an outperformance in the inhibition of BM-TNBC compared with the free drug. Overall, this work provides a promising prospect for the comprehensive treatment of BM-TNBC, which could be generalized to other cell types or used in imaging platforms in the future.
Collapse
Affiliation(s)
- Zimiao Luo
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Shanghai 201203, China,State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Sunyi Wu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Shanghai 201203, China,State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Jianfen Zhou
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Shanghai 201203, China,State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Weixia Xu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Shanghai 201203, China,State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Qianzhu Xu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Shanghai 201203, China,Department of Integrative Medicine, Huashan Hospital, Institutes of Integrative Medicine of Fudan University, Shanghai 200041, China,State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Institutes of Integrative Medicine of Fudan University, Shanghai 200041, China
| | - Cao Xie
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Shanghai 201203, China,State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yu Liu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Shanghai 201203, China,State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Shanghai 201203, China,Department of Integrative Medicine, Huashan Hospital, Institutes of Integrative Medicine of Fudan University, Shanghai 200041, China,Minhang Branch, Zhongshan Hospital and Institute of Fudan-Minghang Academic Health System, Minghang Hospital, Fudan University, Shanghai 201199, China,State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China,Corresponding author.
| |
Collapse
|
23
|
Liu YY, Liu J, Wu H, Zhang Q, Tang XR, Li D, Li CS, Liu Y, Cao A, Wang H. Endocytosis, Distribution, and Exocytosis of Polystyrene Nanoparticles in Human Lung Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 13:nano13010084. [PMID: 36615994 PMCID: PMC9824409 DOI: 10.3390/nano13010084] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 06/13/2023]
Abstract
Nanoplastics, one component of plastic pollution, can enter human bodies via inhalation and thus threaten human health. However, the knowledge about the uptake and exocytosis of nanoplastics in cells of human lung organs is still very limited. Herein, we investigated the endocytosis, distribution, and exocytosis of polystyrene nanoparticles (PS NPs) of 50 nm (G50PS) and 100 nm (R100PS) in A549 cells and BEAS-2B cells. We found that both the cellular uptake of PS NPs increased positively with exposure time and dose, and A549 cells ingested more PS NPs than BEAS-2B cells did. In addition, the intracellular content of G50PS was higher than that of R100PS except at a higher dose and longer time. The ingested PS NPs were distributed mainly in lysosomes, while many G50PS appeared around the cell membrane, and R100PS also accumulated in mitochondria in BEAS-2B cells. As for the exocytosis, R100PS was more difficult to excrete than G50PS. Lysosomes in A549 cells and actin and microtubule in BEAS-2B cells were involved in the exocytosis of the PS NPs. These findings provide detailed information about the translocation of nanoplastics in lung cells, which is valuable for the safety assessment of nanoplastics in the environment.
Collapse
Affiliation(s)
- Yuan-Yuan Liu
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Jie Liu
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Hao Wu
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Qiangqiang Zhang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Xue-Rui Tang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Dan Li
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Chen-Si Li
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Yuanfang Liu
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Aoneng Cao
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Haifang Wang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| |
Collapse
|
24
|
Lu B, Wang J, Scheepers PTJ, Hendriks AJ, Nolte TM. Generic prediction of exocytosis rate constants by size-based surface energies of nanoparticles and cells. Sci Rep 2022; 12:17813. [PMID: 36280701 PMCID: PMC9592603 DOI: 10.1038/s41598-022-20761-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/19/2022] [Indexed: 01/19/2023] Open
Abstract
Nanotechnology brings benefits in fields such as biomedicine but nanoparticles (NPs) may also have adverse health effects. The effects of surface-modified NPs at the cellular level have major implications for both medicine and toxicology. Semi-empirical and mechanism-based models aid to understand the cellular transport of various NPs and its implications for quantitatively biological exposure while avoiding large-scale experiments. We hypothesized relationships between NPs-cellular elimination, surface functionality and elimination pathways by cells. Surface free energy components were used to characterize the transport of NPs onto membranes and with lipid vesicles, covering both influences by size and hydrophobicity of NPs. The model was built based on properties of neutral NPs and cells, defining Van de Waals forces, electrostatic forces and Lewis acid-base (polar) interactions between NPs and vesicles as well as between vesicles and cell membranes. We yielded a generic model for estimating exocytosis rate constants of various neutral NPs by cells based on the vesicle-transported exocytosis pathways. Our results indicate that most models are well fitted (R2 ranging from 0.61 to 0.98) and may provide good predictions of exocytosis rate constants for NPs with differing surface functionalities (prediction errors are within 2 times for macrophages). Exocytosis rates differ between cancerous cells with metastatic potential and non-cancerous cells. Our model provides a reference for cellular elimination of NPs, and intends for medical applications and risk assessment.
Collapse
Affiliation(s)
- Bingqing Lu
- grid.5590.90000000122931605Department of Environmental Science, Institute for Biological and Environmental Sciences, Radboud University Nijmegen, 6500 GL Nijmegen, The Netherlands
| | - Jiaqi Wang
- grid.5590.90000000122931605Department of Environmental Science, Institute for Biological and Environmental Sciences, Radboud University Nijmegen, 6500 GL Nijmegen, The Netherlands
| | - Paul T. J. Scheepers
- grid.5590.90000000122931605Department of Toxicology, Radboud University Nijmegen, 6500 GL Nijmegen, The Netherlands
| | - A. Jan Hendriks
- grid.5590.90000000122931605Department of Environmental Science, Institute for Biological and Environmental Sciences, Radboud University Nijmegen, 6500 GL Nijmegen, The Netherlands
| | - Tom M. Nolte
- grid.5590.90000000122931605Department of Environmental Science, Institute for Biological and Environmental Sciences, Radboud University Nijmegen, 6500 GL Nijmegen, The Netherlands
| |
Collapse
|
25
|
Liu YY, Sun ZX, Liu J, Zhang Q, Liu Y, Cao A, Sun YP, Wang H. On the Cellular Uptake and Exocytosis of Carbon Dots─Significant Cell Type Dependence and Effects of Cell Division. ACS APPLIED BIO MATERIALS 2022; 5:4378-4389. [PMID: 36044400 DOI: 10.1021/acsabm.2c00542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Understanding the cellular uptake and exocytosis processes of nanoparticles (NPs) is essential for developing the nanomedicines and assessing the health risk of nanomaterials. Considerable efforts have been made to reveal how physicochemical properties of NPs influence these processes. However, little attention has been paid to how cell type impacts these processes, especially exocytosis. Herein, the uptake and exocytosis of the carbon dots (CDs) obtained from the carbonization of citric acid with polyethylenimine (PEI) oligomers (CDs-PEI) in five human cell lines (HeLa, A549, BEAS-2B, A431, and MDA-MB-468) are analyzed to understand how cell type influences the fate of CDs in cells. The cell division is taken into account by the correction of cell number for accurate quantification of the uptake and exocytosis of CDs-PEI. The results indicate that the cell type significantly affects the cellular uptake, trafficking, and exocytosis of CDs-PEI. Among the cell types investigated, MDA-MB-468 cells have the greatest capacity for both uptake and exocytosis, and HeLa cells have the least capacity. The kinetics of the exocytosis largely follows a single exponential decay function, with the remaining CDs-PEI in cells reaching plateaus within 24 h. The kinetic parameters are cell-dependent but insensitive to the initial intracellular CDs-PEI content. Generally, the Golgi apparatus pathways are more important in exocytosis than the lysosomal pathway, and the locations of CDs-PEI in the beginning of exocytosis are not correlated with their exocytosis pathways. The findings on the cell type-dependent cellular uptake and exocytosis reported here may be valuable to the future design of high-performance and safe CDs and related nanomaterials in general.
Collapse
Affiliation(s)
- Yuan-Yuan Liu
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Zao-Xia Sun
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Jie Liu
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Qiangqiang Zhang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Yuanfang Liu
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Aoneng Cao
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Ya-Ping Sun
- Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
| | - Haifang Wang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| |
Collapse
|
26
|
TRPML3 enhances drug resistance in non-small cell lung cancer cells by promoting Ca 2+-mediated lysosomal trafficking. Biochem Biophys Res Commun 2022; 627:152-159. [PMID: 36037747 DOI: 10.1016/j.bbrc.2022.08.051] [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: 08/08/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/23/2022]
Abstract
Lysosomes are emerging as versatile signaling hubs that mediate numerous cellular processes, including the development of drug resistance in cancer cells. Transient receptor potential mucolipin 3 (TRPML3), an endolysosomal Ca2+-permeable channel, is implicated in regulating lysosomal trafficking during endocytosis and autophagy. However, the role of TRPML3 in cancer progression remains unclear. In this study, we focused on identifying key molecules that modulate exosomal release triggered by lysosomal exocytosis during the development of gefitinib resistance in non-small cell lung cancer (NSCLC). We found that the basal release of exosomes and lysosomal exocytosis is higher in the gefitinib-resistant NSCLC cell line HCC827/GR than in the gefitinib-sensitive NSCLC cell line HCC827. Notably, exosomal release and lysosomal exocytosis were associated with an increase in TRPML3 expression. Lysosomal Ca2+ release via TRPML3 was triggered by the gefitinib-mediated elevation of lysosomal pH. Furthermore, TRPML3 deficiency enhanced the gefitinib-mediated increase in sub-G0 cell population, reduction of cell proliferation, and poly (ADP-ribose) polymerase cleavage. These data demonstrated that TRPML3 is a promising modulator of drug resistance. By sensing the elevation of lysosomal pH, it mediates lysosomal Ca2+ release, lysosomal trafficking and exocytosis, and exosomal release. Taken together, our study is the first to report the autonomous defense mechanism developed in NSCLC cells against the small-molecule tyrosine kinase inhibitor gefitinib, leading to acquired drug resistance.
Collapse
|
27
|
Size-dependent placental retention effect of liposomes in ICR pregnant mice: Potential superiority in placenta-derived disease therapy. Int J Pharm 2022; 625:122121. [PMID: 35987320 DOI: 10.1016/j.ijpharm.2022.122121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 08/10/2022] [Accepted: 08/13/2022] [Indexed: 11/23/2022]
Abstract
The great challenge in developing safe medications for placenta-derived diseases is to reduce or eliminate fetal drug exposure while still providing the necessary therapeutic effect. Rapid advances in nanotechnology have brought opportunities for the therapy of placenta-derived disease through accumulating the drug in the placenta while reducing its placental penetration. Among various nanocarriers, liposomes are regarded as an ideal type of carrier for placental drug delivery due to their biosafety and biodegradability. However, their placental retention effect with different particle sizes has not been studied. This research aimed to explore a suitable size of liposomes for placenta drug delivery. Cy 5 dye was chosen as a model molecule for tracing the distribution of three different-sized liposomes (∼80 nm, 200 nm, and 500 nm) in ICR pregnant mice. The stability, cytotoxicity, and cellular uptake study of Cy 5-loaded liposomes were performed. The in vivo fluorescence studies on ICR pregnant mice suggested that the particle size of liposomes was positively correlated with the degree of liposome aggregation in the placenta. The ratio of fluorescence in the placenta and fetus section (P/F value) was proposed to evaluate the placental retention effect of different-sized liposomes. The results showed that the liposomes with 500 nm had the highest P/F value and thus exhibited the strongest placental retention effect and the weakest placental penetration ability. Moreover, liquid chromatography-mass spectrometry analysis confirmed the reliability of the fluorescence section analysis in exploring the placental retention effect of nanovehicles. In general, this study introduced a simple and intuitive method to evaluate the placental retention effect of nanoplatforms and defined a suitable size of liposomes for placenta-derived disease drug delivery.
Collapse
|
28
|
Ma Z, Wong S, Forgham H, Esser L, Lai M, Leiske M, Kempe K, Sharbeen G, Youkhana J, Mansfeld F, Quinn J, Phillips P, Davis T, Kavallaris M, McCarroll J. Aerosol delivery of star polymer-siRNA nanoparticles as a therapeutic strategy to inhibit lung tumor growth. Biomaterials 2022; 285:121539. [DOI: 10.1016/j.biomaterials.2022.121539] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 01/12/2023]
|
29
|
Zhang M, Kim DS, Patel R, Wu Q, Kim K. Intracellular Trafficking and Distribution of Cd and InP Quantum Dots in HeLa and ML-1 Thyroid Cancer Cells. NANOMATERIALS 2022; 12:nano12091517. [PMID: 35564224 PMCID: PMC9104504 DOI: 10.3390/nano12091517] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 02/01/2023]
Abstract
The study of the interaction of engineered nanoparticles, including quantum dots (QDs), with cellular constituents and the kinetics of their localization and transport, has provided new insights into their biological consequences in cancers and for the development of effective cancer therapies. The present study aims to elucidate the toxicity and intracellular transport kinetics of CdSe/ZnS and InP/ZnS QDs in late-stage ML-1 thyroid cancer using well-tested HeLa as a control. Our XTT (2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide) viability assay (Cell Proliferation Kit II) showed that ML-1 cells and non-cancerous mouse fibroblast cells exhibit no viability defect in response to these QDs, whereas HeLa cell viability decreases. These results suggest that HeLa cells are more sensitive to the QDs compared to ML-1 cells. To test the possibility that transporting rates of QDs are different between HeLa and ML-1 cells, we performed a QD subcellular localization assay by determining Pearson’s Coefficient values and found that HeLa cells showed faster QDs transporting towards the lysosome. Consistently, the ICP-OES test showed the uptake of CdSe/ZnS QDs in HeLa cells was significantly higher than in ML-1 cells. Together, we conclude that high levels of toxicity in HeLa are positively correlated with the traffic rate of QDs in the treated cells.
Collapse
Affiliation(s)
- Min Zhang
- Department of Biology, Missouri State University, 901 S National, Springfield, MO 65897, USA;
| | - Daniel S. Kim
- Emory College of Arts and Science, Emory University, 201 Dowman Dr., Atlanta, GA 30322, USA;
| | - Rishi Patel
- Jordan Valley Innovation Center, Missouri State University, 542 N Boonville Ave, Springfield, MO 65806, USA; (R.P.); (Q.W.)
| | - Qihua Wu
- Jordan Valley Innovation Center, Missouri State University, 542 N Boonville Ave, Springfield, MO 65806, USA; (R.P.); (Q.W.)
| | - Kyoungtae Kim
- Department of Biology, Missouri State University, 901 S National, Springfield, MO 65897, USA;
- Correspondence: ; Tel.: +1-417-836-5440; Fax: +1-417-836-5126
| |
Collapse
|
30
|
Tumor Tropic Delivery of Hyaluronic Acid-Poly (D,L-lactide-co-glycolide) Polymeric Micelles Using Mesenchymal Stem Cells for Glioma Therapy. Molecules 2022; 27:molecules27082419. [PMID: 35458619 PMCID: PMC9027425 DOI: 10.3390/molecules27082419] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 03/31/2022] [Accepted: 04/07/2022] [Indexed: 01/13/2023] Open
Abstract
Tumor penetration and the accumulation of nanomedicines are crucial challenges in solid tumor therapy. By taking advantage of the MSC tumor-tropic property, we developed a mesenchymal stem cell (MSC)-based drug delivery system in which paclitaxel (PTX)-encapsulating hyaluronic acid-poly (D,L-lactide-co-glycolide) polymeric micelles (PTX/HA-PLGA micelles) were loaded for glioma therapy. The results indicated that CD44 overexpressed on the surface of both MSCs and tumor cells not only improved PTX/HA-PLGA micelle loading in MSCs, but also promoted the drug transfer between MSCs and adjacent cancer cells. It was hypothesized that CD44-mediated transcytosis played a crucial role and allowed deep glioma penetration depending on sequential intra–intercellular delivery via endocytosis–exocytosis. MSC-micelles were able to infiltrate from normal brain parenchyma towards contralateral tumors and led to the eradication of glioma. The survival of orthotopic glioma-bearing rats was significantly extended. In conclusion, the MSC-based delivery of HA-PLGA micelles is a potential strategy for tumor-targeting drug delivery.
Collapse
|
31
|
Kim IY, Kwak M, Kim J, Lee TG, Heo MB. Comparative Study on Nanotoxicity in Human Primary and Cancer Cells. NANOMATERIALS 2022; 12:nano12060993. [PMID: 35335806 PMCID: PMC8955245 DOI: 10.3390/nano12060993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 02/05/2023]
Abstract
Nanomaterial toxicity tests using normal and cancer cells may yield markedly different results. Here, nanomaterial toxicity between cancer and primary human cells was compared to determine the basic cell line selection criteria for nanomaterial toxicity analyses. Specifically, we exposed two cancer (A549 and HepG2) and two normal cell lines (NHBE and HH) cell lines to SiO2 nanoparticles (NPs) and evaluated the cytotoxicity (MTS assay), cell death mode, and intracellular NP retention. MTS assay results revealed higher sensitivity of HH cells to SiO2 NPs than HepG2 cells, while no difference was observed between NHBE and A549 cells. In addition, SiO2 NPs primarily induced necrosis in all the cell lines. Moreover, we evaluated NP accumulation by treating the cell lines with fluorescein-isothiocyanate-labeled SiO2 NPs. After 48 h of treatment, less than 10% of A549 and HepG2 cells and more than 30% of NHBE and HH cells contained the labeled NPs. Collectively, our results suggest that cell viability, death mode, and intracellular compound accumulation could be assessed using cancer cells. However, the outcomes of certain investigations, such as intracellular NP retention, may differ between cancer and normal cells.
Collapse
|
32
|
Ho LWC, Chan CKW, Han R, Lau YFY, Li H, Ho YP, Zhuang X, Choi CHJ. Mammalian Cells Exocytose Alkylated Gold Nanoparticles via Extracellular Vesicles. ACS NANO 2022; 16:2032-2045. [PMID: 35137580 DOI: 10.1021/acsnano.1c07418] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Understanding the exocytosis of nanoparticles (NPs) from cells is valuable because it informs design rules of NPs that support desirable cellular retention for nanomedicine applications, but investigations into the mechanism for the exocytosis of NPs remain scarce. We elucidate the mechanism for the exocytosis of dodecyl-terminated, polyethylene glycol-coated gold NPs (termed "dodecyl-PEG-AuNP"). The Au core enables ultrastructural differentiation of the exocytosed NPs from the nearby extracellular vesicles (EVs). The PEG shell prevents interparticle agglomeration or aggregation that disfavors exocytosis. The minute amounts of alkyl chains on the PEG shell not only promote cellular uptake but also improve exocytosis by up to 4-fold higher probability and upregulate exocytosis- and vesicle-related genes. After entering Kera-308 keratinocytes and trafficking to multivesicular bodies and lysosomes, these NPs exit the cell predominantly via unconventional exocytosis, accompanied by enhanced secretion of sub-100 nm, CD81-enriched exosomes. The pathway for NP exocytosis and subpopulation of EVs that are secreted alongside the exocytosed NPs depends on dodecyl loading. This work provides insights into dissecting the mechanism of NP exocytosis and its relationship with EV secretion.
Collapse
|
33
|
Shi M, Jiang Z, Xiao Y, Song Y, Tang R, Zhang L, Huang J, Tian Y, Zhou S. Stapling of short cell-penetrating peptides for enhanced tumor cell-and-tissue dual-penetration. Chem Commun (Camb) 2022; 58:2299-2302. [PMID: 35075473 DOI: 10.1039/d1cc06595e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Effective delivery of therapeutics to tumors is generally hampered by the limited penetration of biological barriers imposed by the tumor microenvironment. Despite the broad applications of cell-penetrating peptides (CPPs) for intracellular delivery of therapeutics across membrane bilayers, the discovery of novel CPPs with enhanced tumor tissue permeability remains largely unexplored. Herein, we identified two short stapled CPPs with aromatic cross-links that confer superior dual-penetration in tumor cells and tissues over their linear counterparts. This work may benefit the future applications of constrained CPPs as powerful molecular transporters to access deeper tumor tissues.
Collapse
Affiliation(s)
- Mengzhen Shi
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, P. R. China.
| | - Zherui Jiang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, P. R. China.
| | - Yao Xiao
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, P. R. China.
| | - Yue Song
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, P. R. China.
| | - Rui Tang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, P. R. China.
| | - Ling Zhang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, P. R. China.
| | - Jianfeng Huang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, P. R. China.
| | - Yuan Tian
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, P. R. China.
| | - Shaobing Zhou
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, P. R. China.
| |
Collapse
|
34
|
Van de Vyver T, De Smedt SC, Raemdonck K. Modulating intracellular pathways to improve non-viral delivery of RNA therapeutics. Adv Drug Deliv Rev 2022; 181:114041. [PMID: 34763002 DOI: 10.1016/j.addr.2021.114041] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/12/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022]
Abstract
RNA therapeutics (e.g. siRNA, oligonucleotides, mRNA, etc.) show great potential for the treatment of a myriad of diseases. However, to reach their site of action in the cytosol or nucleus of target cells, multiple intra- and extracellular barriers have to be surmounted. Several non-viral delivery systems, such as nanoparticles and conjugates, have been successfully developed to meet this requirement. Unfortunately, despite these clear advances, state-of-the-art delivery agents still suffer from relatively low intracellular delivery efficiencies. Notably, our current understanding of the intracellular delivery process is largely oversimplified. Gaining mechanistic insight into how RNA formulations are processed by cells will fuel rational design of the next generation of delivery carriers. In addition, identifying which intracellular pathways contribute to productive RNA delivery could provide opportunities to boost the delivery performance of existing nanoformulations. In this review, we discuss both established as well as emerging techniques that can be used to assess the impact of different intracellular barriers on RNA transfection performance. Next, we highlight how several modulators, including small molecules but also genetic perturbation technologies, can boost RNA delivery by intervening at differing stages of the intracellular delivery process, such as cellular uptake, intracellular trafficking, endosomal escape, autophagy and exocytosis.
Collapse
Affiliation(s)
- Thijs Van de Vyver
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Koen Raemdonck
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
35
|
Wang X, Wang WX. Cu-based nanoparticle toxicity to zebrafish cells regulated by cellular discharges. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 292:118296. [PMID: 34627961 DOI: 10.1016/j.envpol.2021.118296] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/17/2021] [Accepted: 10/04/2021] [Indexed: 05/21/2023]
Abstract
Cellular transport of metal nanoparticles (NPs) is critical in determining their potential toxicity, but the transformation of metal ions released from the internalized NPs is largely unknown. Cu-based NPs are the only metallic-based NPs that are reported to induce higher toxicity compared with their corresponding ions, likely due to their unique cellular turnover. In the present study, we developed a novel gold core to differentiate the particulate and ionic Cu in the Cu2O microparticles (MPs), and the kinetics of bioaccumulation, exocytosis, and cytotoxicity of Au@Cu2O MPs to zebrafish embryonic cells were subsequently studied. We demonstrated that the internalized MPs were rapidly dissolved to Cu ions, which then undergo lysosome-mediated exocytosis. The uptake rate of smaller MPs (130 nm) was lower than that of larger ones (200 nm), but smaller MPs were dissolved much quickly in cells and therefore activated the exocytosis more quickly. The rapid release of Cu ions resulted in an immediate toxic action of Cu2O MPs, while the cell deaths mainly occurred by necrosis. During this process, the buffering ability of glutathione greatly alleviated the Cu toxicity. Therefore, although the turnover of intracellular Cu at a sublethal exposure level was hundred times faster than the basal values, labile Cu(I) concentration increased by only 2 times at most. Overall, this work provided new insights into the toxicity of copper NPs, suggesting that tolerance to Cu-based NPs depended on their ability to discharge the released Cu ions.
Collapse
Affiliation(s)
- Xiangrui Wang
- School of Energy and Environment, Hong Kong Branch of the Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China; Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen, 518057, China
| | - Wen-Xiong Wang
- School of Energy and Environment, Hong Kong Branch of the Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong Kong, China; Research Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen, 518057, China.
| |
Collapse
|
36
|
Hadji H, Bouchemal K. Effect of micro- and nanoparticle shape on biological processes. J Control Release 2021; 342:93-110. [PMID: 34973308 DOI: 10.1016/j.jconrel.2021.12.032] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 12/15/2022]
Abstract
In the drug delivery field, there is beyond doubt that the shape of micro- and nanoparticles (M&NPs) critically affects their biological fate. Herein, following an introduction describing recent technological advances for designing nonspherical M&NPs, we highlight the role of particle shape in cell capture, subcellular distribution, intracellular drug delivery, and cytotoxicity. Then, we discuss theoretical approaches for understanding the effect of particle shape on internalization by the cell membrane. Subsequently, recent advances on shape-dependent behaviors of M&NPs in the systemic circulation are detailed. In particular, the interaction of M&NPs with blood proteins, biodistribution, and circulation under flow conditions are analyzed. Finally, the hurdles and future directions for developing nonspherical M&NPs are underscored.
Collapse
Affiliation(s)
- Hicheme Hadji
- Université Paris-Saclay, Institut Galien Paris Saclay, CNRS UMR 8612, 92296 Châtenay-Malabry, France
| | - Kawthar Bouchemal
- Université Paris-Saclay, Institut Galien Paris Saclay, CNRS UMR 8612, 92296 Châtenay-Malabry, France.
| |
Collapse
|
37
|
Liu H, Deng S, Han L, Ren Y, Gu J, He L, Liu T, Yuan ZX. Mesenchymal stem cells, exosomes and exosome-mimics as smart drug carriers for targeted cancer therapy. Colloids Surf B Biointerfaces 2021; 209:112163. [PMID: 34736220 DOI: 10.1016/j.colsurfb.2021.112163] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 10/10/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent stem cells with the capacity to differentiate into several cell types under appropriate conditions. They also possess remarkable antitumor features that make them a novel choice to treat cancers. Accumulating evidence suggest that the MSCs-derived extracellular vesicles, known as exosomes, play an essential role in the therapeutic effects of MSCs mainly by carrying biologically active factors. However, limitations such as low yield of exosomes and difficulty in isolation and purification hinder their clinical applications. To overcome these issues, research on development of exosome-mimics has attracted great attention. This systematic review represents, to the best of our knowledge, the first thorough evaluations of the innate antineoplastic features of MSCs-derived exosomes or exosome-mimics, the methods of drug loading, application as drug delivery system and their impacts on targeted cancer therapy. Importantly, we dissect the commonalities and differences as well as address the shortcomings of work accumulated over the last two decades and discuss how this information can serve as a guide map for optimal experimental design implementation ultimately aiding the effective transition into clinical trials.
Collapse
Affiliation(s)
- Hongmei Liu
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China
| | - Shichen Deng
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu, Sichuan, China
| | - Lu Han
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China
| | - Yan Ren
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China
| | - Jian Gu
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China
| | - Lili He
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China.
| | - Tianqing Liu
- NICM Health Research Institute, Western Sydney University, Westmead, Australia.
| | - Zhi-Xiang Yuan
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China.
| |
Collapse
|
38
|
Liu YY, Chang Q, Sun ZX, Liu J, Deng X, Liu Y, Cao A, Wang H. Fate of CdSe/ZnS quantum dots in cells: Endocytosis, translocation and exocytosis. Colloids Surf B Biointerfaces 2021; 208:112140. [PMID: 34597939 DOI: 10.1016/j.colsurfb.2021.112140] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/08/2021] [Accepted: 09/23/2021] [Indexed: 11/29/2022]
Abstract
Semiconductor quantum dots (QDs) have been extensively explored for extensive bioapplications, yet their cellular fate, especially exocytosis, has not been thoroughly investigated. Herein, we systematically investigated the whole cellular process from the endocytosis, intercellular trafficking, to the exocytosis of a typical QD, core/shell CdSe/ZnS QD. Using confocal laser scanning microscopy and flow cytometry, and after carefully eliminating the effect of cell division, we found that the QDs were internalized by HeLa cells with a time-, dose-, and serum-dependent manner. The cellular uptake was inhibited by serum, but eventually peaked after 4-6 h incubation with or without serum. The primary endocytosis pathway was clathrin-mediated, and actin- and microtubule-dependent in the medium with serum, while the caveolae-mediated endocytosis and macropinocytosis were more important for the QDs in the serum-free medium. Inside cells, most QDs distributed in lysosomes, and some entered mitochondria, endoplasmic reticulum, and Golgi apparatus. The translocation of the QDs from other organelles to Golgi apparatus was observed. The exocytosis of QDs was faster than the endocytosis, reaching the maximum in about one hour after cultured in fresh culture medium, with around 60% of the internalized QDs remained undischarged. The exocytosis process was energy- and actin-dependent, and the lysosome exocytosis and endoplasmic reticulum/Golgi pathway were the main routes. This study provides a full picture of behavior and fate of QDs in cells, which may facilitate the design of ideal QDs applied in biomedical and other fields.
Collapse
Affiliation(s)
- Yuan-Yuan Liu
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Qing Chang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Zao-Xia Sun
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Jie Liu
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Xiaoyong Deng
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Yuanfang Liu
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China; Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Aoneng Cao
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China.
| | - Haifang Wang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
39
|
Xu K, Basu N, George S. Dietary nanoparticles compromise epithelial integrity and enhance translocation and antigenicity of milk proteins: An in vitro investigation. NANOIMPACT 2021; 24:100369. [PMID: 35559811 DOI: 10.1016/j.impact.2021.100369] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/24/2021] [Accepted: 11/18/2021] [Indexed: 05/28/2023]
Abstract
Nanoparticles (NPs) are increasingly being used in the food sector, yet little is known about the potential health risks associated with oral exposure to dietary NPs. In this study, the most widely used NPs in food industry including food grade silicon dioxide (SiO2), titanium dioxide (TiO2) and silver (Ag), along with their non-food grade and bulk counterparts, are characterized for physicochemical properties and molecular, cellular, and intracellular effects on human intestinal epithelial cells (Caco-2 and HIEC-6). Silver NPs are the most cytotoxic and induce significant cellular changes in oxidative stress, Ca2+ flux and mitochondria function, leading to cellular junction disruption at the lowest exposure concentration. At higher testing concentrations, NPs but not microparticles of SiO2 and TiO2 cause sublethal cellular responses and remodel tight junctions without impairing epithelial integrity. To relate the cellular results to key events in GI disorder progression, NPs are exposed to an in vitro co-culture model for cow's milk allergy comprised of Caco-2 and allergy sera-primed mast cells (LUVA). All particle treatments increase the allergen delivery across intestinal epithelium and subsequent allergy responses. Overall, the study has identified a particle-dependent alteration in intestinal epithelium and highlighted potential safety concerns of dietary NPs.
Collapse
Affiliation(s)
- Ke Xu
- Department of Food Science and Agricultural Chemistry, McGill University, 21111 Lakeshore Dr, Sainte-Anne-de-Bellevue, QC H9X, Canada
| | - Niladri Basu
- Department of Natural Resource Science, McGill University, 21111 Lakeshore Dr, Sainte-Anne-de-Bellevue, QC H9X, Canada
| | - Saji George
- Department of Food Science and Agricultural Chemistry, McGill University, 21111 Lakeshore Dr, Sainte-Anne-de-Bellevue, QC H9X, Canada.
| |
Collapse
|
40
|
Saha T, Mondal J, Khiste S, Lusic H, Hu ZW, Jayabalan R, Hodgetts KJ, Jang H, Sengupta S, Lee SE, Park Y, Lee LP, Goldman A. Nanotherapeutic approaches to overcome distinct drug resistance barriers in models of breast cancer. NANOPHOTONICS 2021; 10:3063-3073. [PMID: 34589378 PMCID: PMC8478290 DOI: 10.1515/nanoph-2021-0142] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Targeted delivery of drugs to tumor cells, which circumvent resistance mechanisms and induce cell killing, is a lingering challenge that requires innovative solutions. Here, we provide two bioengineered strategies in which nanotechnology is blended with cancer medicine to preferentially target distinct mechanisms of drug resistance. In the first 'case study', we demonstrate the use of lipid-drug conjugates that target molecular signaling pathways, which result from taxane-induced drug tolerance via cell surface lipid raft accumulations. Through a small molecule drug screen, we identify a kinase inhibitor that optimally destroys drug tolerant cancer cells and conjugate it to a rationally-chosen lipid scaffold, which enhances anticancer efficacy in vitro and in vivo. In the second 'case study', we address resistance mechanisms that can occur through exocytosis of nanomedicines. Using adenocarcinoma HeLa and MCF-7 cells, we describe the use of gold nanorod and nanoporous vehicles integrated with an optical antenna for on-demand, photoactivation at ~650 nm enabling release of payloads into cells including cytotoxic anthracyclines. Together, these provide two approaches, which exploit engineering strategies capable of circumventing distinct resistance barriers and induce killing by multimodal, including nanophotonic mechanisms.
Collapse
Affiliation(s)
- Tanmoy Saha
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jayanta Mondal
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Sachin Khiste
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Hrvoje Lusic
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Zhang-Wei Hu
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | | | | | - HaeLin Jang
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Shiladitya Sengupta
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Somin Eunice Lee
- Department of Electrical & Computer Engineering, University of Michigan, Ann Arbor, MI48109,USA
- Department of Biomedical Engineering, Biointerfaces Institute, Applied Physics, Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI48109,USA
| | - Younggeun Park
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI48109,USA
| | - Luke P. Lee
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Aaron Goldman
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cancer Immunology, Dana Farber/Harvard Cancer Center, Boston, MA, USA
| |
Collapse
|
41
|
Chen S, Zhong Y, Fan W, Xiang J, Wang G, Zhou Q, Wang J, Geng Y, Sun R, Zhang Z, Piao Y, Wang J, Zhuo J, Cong H, Jiang H, Ling J, Li Z, Yang D, Yao X, Xu X, Zhou Z, Tang J, Shen Y. Enhanced tumour penetration and prolonged circulation in blood of polyzwitterion-drug conjugates with cell-membrane affinity. Nat Biomed Eng 2021; 5:1019-1037. [PMID: 33859387 DOI: 10.1038/s41551-021-00701-4] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 02/16/2021] [Indexed: 02/01/2023]
Abstract
Effective anticancer nanomedicines need to exhibit prolonged circulation in blood, to extravasate and accumulate in tumours, and to be taken up by tumour cells. These contrasting criteria for persistent circulation and cell-membrane affinity have often led to complex nanoparticle designs with hampered clinical translatability. Here, we show that conjugates of small-molecule anticancer drugs with the polyzwitterion poly(2-(N-oxide-N,N-diethylamino)ethyl methacrylate) have long blood-circulation half-lives and bind reversibly to cell membranes, owing to the negligible interaction of the polyzwitterion with proteins and its weak interaction with phospholipids. Adsorption of the polyzwitterion-drug conjugates to tumour endothelial cells and then to cancer cells favoured their transcytosis-mediated extravasation into tumour interstitium and infiltration into tumours, and led to the eradication of large tumours and patient-derived tumour xenografts in mice. The simplicity and potency of the polyzwitterion-drug conjugates should facilitate the design of translational anticancer nanomedicines.
Collapse
Affiliation(s)
- Siqin Chen
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Yin Zhong
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Wufa Fan
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China.,Department of Polymer Science and Engineering, Peking University, Beijing, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Guowei Wang
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Quan Zhou
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Jinqiang Wang
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Yu Geng
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Rui Sun
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Zhen Zhang
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Jianguo Wang
- Department of Surgery, First Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianyong Zhuo
- Department of Surgery, First Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Hailin Cong
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, China
| | - Haiping Jiang
- Department of Medical Oncology, The First Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Jun Ling
- Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Zichen Li
- Department of Polymer Science and Engineering, Peking University, Beijing, China
| | - Dingding Yang
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xin Yao
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xiao Xu
- Department of Surgery, First Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China. .,Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China. .,Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China.
| |
Collapse
|
42
|
He B, Sui X, Yu B, Wang S, Shen Y, Cong H. Recent advances in drug delivery systems for enhancing drug penetration into tumors. Drug Deliv 2021; 27:1474-1490. [PMID: 33100061 PMCID: PMC7594734 DOI: 10.1080/10717544.2020.1831106] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The emergence of nanomaterials for drug delivery provides the opportunity to avoid the side effects of systemic drug administration and injury caused by the removal of tumors, delivering great promise for future cancer treatments. However, the efficacy of current nano drugs is not significantly better than that of the original drug treatments. The important reason is that nano drugs enter the tumor vasculature, remaining close to the blood vessels and unable to enter the tumor tissue or tumor cells to complete the drug delivery process. The low efficiency of drug penetration into tumors has become a bottleneck restricting the development of nano-drugs. Herein, we present a systematic overview of recent advances on the design of nano-drug carriers in drug delivery systems for enhancing drug penetration into tumors. The review is organized into four sections: The drug penetration process in tumor tissue includes paracellular and transcellular transport, which is summarized first. Strategies that promote tumor penetration are then introduced, including methods of remodeling the tumor microenvironment, charge inversion, dimensional change, and surface modification of ligands which promote tissue penetration. Conclusion and the prospects for the future development of drug penetration are finally briefly illustrated. The review is intended to provide thoughts for effective treatment of cancer by summarizing strategies for promoting the endocytosis of nano drugs into tumor cells.
Collapse
Affiliation(s)
- Bin He
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Xin Sui
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Bing Yu
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.,Key Laboratory of Bio-Fibers and Eco-Textiles, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, China
| | - Song Wang
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Youqing Shen
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.,Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Hailin Cong
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.,Key Laboratory of Bio-Fibers and Eco-Textiles, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, China
| |
Collapse
|
43
|
Tieu T, Wei Y, Cifuentes‐Rius A, Voelcker NH. Overcoming Barriers: Clinical Translation of siRNA Nanomedicines. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100108] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Terence Tieu
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
- CSIRO Manufacturing Bayview Avenue Clayton VIC 3168 Australia
| | - Yingkai Wei
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
| | - Anna Cifuentes‐Rius
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
| | - Nicolas H. Voelcker
- Parkville Campus 381 Royal Parade Monash Institute of Pharmaceutical Sciences Monash University Parkville VIC 3052 Australia
- CSIRO Manufacturing Bayview Avenue Clayton VIC 3168 Australia
- Melbourne Centre for Nanofabrication 151 Wellington Road Victorian Node of the Australian National Fabrication Facility Clayton VIC 3168 Australia
| |
Collapse
|
44
|
Liu L, Xu K, Zhang B, Ye Y, Zhang Q, Jiang W. Cellular internalization and release of polystyrene microplastics and nanoplastics. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 779:146523. [PMID: 34030247 DOI: 10.1016/j.scitotenv.2021.146523] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/12/2021] [Accepted: 03/12/2021] [Indexed: 06/12/2023]
Abstract
Microplastics and nanoplastics can accumulate in organisms after being ingested, be transported in the food web, and ultimately threaten human health. An understanding of the cellular internalization and release of micro(nano)plastics is important to predict their cytotoxicity. In this study, 50 nm, 500 nm and 5 μm polystyrene particles (PS50, PS500 and PS5000) were exposed to both model cell membranes and rat basophilic leukemia (RBL-2H3) cells. PS50 and PS500 absorb on the model membrane due to hydrophobic interactions and Van der Waals' forces, and may also penetrate the model membrane. PS50 and PS500 are internalized into living cells via both passive membrane penetration and active endocytosis. The passive membrane penetration is due to the partition of polystyrene particles in the water-phospholipid system. The endocytosis of PS50 occurs through the clathrin-mediated pathway, the caveolin-mediated pathway and macropinocytosis, but endocytosis of PS500 is mainly via the macropinocytosis. PS5000 cannot adhere to the cell membrane or be internalized into cells due to its large size and weak Brownian motion. The endocytosed PS50 and PS500 mainly accumulate in the lysosomes. The passively internalized PS50 and PS500 initially distribute in the cytoplasm not in lysosomes, but are transported to lysosomes with energy supply. PS50 and PS500 are excreted from cells via energy-free penetration and energy-dependent lysosomal exocytosis. The masses of the internalized PS50 inside the cells and the excreted PS50 outside the cells were both higher than the masses of PS500, indicating that the smaller particles are more easily enter or leave cells than do their larger counterparts. Our findings will contribute to the risk assessment of micro(nano)plastics and their safe application.
Collapse
Affiliation(s)
- Ling Liu
- Environment Research Institute, Shandong University, Qingdao 266237, China
| | - Kexin Xu
- Environment Research Institute, Shandong University, Qingdao 266237, China
| | - Bowen Zhang
- Environment Research Institute, Shandong University, Qingdao 266237, China
| | - Yiyuan Ye
- Environment Research Institute, Shandong University, Qingdao 266237, China
| | - Qiu Zhang
- School of Environmental Sciences and Engineering, Shandong University, Qingdao 266237, China
| | - Wei Jiang
- Environment Research Institute, Shandong University, Qingdao 266237, China.
| |
Collapse
|
45
|
Direct in vivo reprogramming with non-viral sequential targeting nanoparticles promotes cardiac regeneration. Biomaterials 2021; 276:121028. [PMID: 34293701 DOI: 10.1016/j.biomaterials.2021.121028] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/23/2021] [Accepted: 07/12/2021] [Indexed: 12/11/2022]
Abstract
microRNA-mediated direct cardiac reprogramming, directly converts fibroblasts into induced cardiomyocyte-like cells (iCMs), which holds great promise in cardiac regeneration therapy. However, effective approaches to deliver therapeutic microRNA into cardiac fibroblasts (CFs) to induce in vivo cardiac reprogramming remain to be explored. Herein, a non-viral biomimetic system to directly reprogram CFs for cardiac regeneration after myocardial injury was developed by coating FH peptide-modified neutrophil-mimicking membranes on mesoporous silicon nanoparticles (MSNs) loaded with microRNA1, 133, 208, and 499 (miR Combo). Through utilizing the natural inflammation-homing ability of neutrophil membrane protein and FH peptide's high affinity to tenascin-C (TN-C) produced by CFs, this nanoparticle could realize sequential targeting to CFs in the injured heart and precise intracellular delivery of miRCombo, which induced reprogramming resident CFs into iCMs. In a mouse model of myocardial ischemia/reperfusion injury, intravenous injection of the nanoparticles successfully delivered miRCombo into fibroblasts and led to efficient reprogramming, resulting in improved cardiac function and attenuated fibrosis. This delivery system is minimally invasive and bio-safe, providing a proof-of-concept for biomimetic and sequential targeting nanomedicine delivery system for microRNA-mediated reprogramming therapy in multiple diseases.
Collapse
|
46
|
Kamali Shahri SM, Sharifi S, Mahmoudi M. Interdependency of influential parameters in therapeutic nanomedicine. Expert Opin Drug Deliv 2021; 18:1379-1394. [PMID: 33887999 DOI: 10.1080/17425247.2021.1921732] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction:Current challenges to successful clinical translation of therapeutic nanomedicine have discouraged many stakeholders, including patients. Significant effort has been devoted to uncovering the reasons behind the less-than-expected success, beyond failures or ineffectiveness, of therapeutic nanomedicine products (e.g. cancer nanomedicine). Until we understand and address the factors that limit the safety and efficacy of NPs, both individually and in combination, successful clinical development will lag.Areas covered:This review highlights the critical roles of interdependent factors affecting the safety and therapeutic efficacy of therapeutic NPs for drug delivery applications.Expert opinion:Deep analysis of the current nanomedical literature reveals ahistory of unanticipated complexity by awide range of stakeholders including researchers. In the manufacture of nanomedicines themselves, there have been persistent difficulties with reproducibility and batch-to-batch variation. The unanticipated complexity and interdependency of nano-bio parameters has delayed our recognition of important factors affecting the safety and therapeutic efficacy of nanomedicine products. These missteps have had many factors including our lack of understanding of the interdependency of various factors affecting the biological identity and fate of NPs and biased interpretation of data. All these issues could raise significant concern regarding the reproducibility- or even the validity- of past publications that in turn formed the basis of many clinical trials of therapeutic nanomedicines. Therefore, the individual and combined effects of previously overlooked factors on the safety and therapeutic efficacy of NPs need to be fully considered in nanomedicine reports and product development.
Collapse
Affiliation(s)
- Seyed Mehdi Kamali Shahri
- Department of Radiology and Precision Health Program, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Shahriar Sharifi
- Department of Radiology and Precision Health Program, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Morteza Mahmoudi
- Department of Radiology and Precision Health Program, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
47
|
Khan MA, Fugate M, Rogers DT, Sambi J, Littleton JM, Rankin SE, Knutson BL. Mechanism of Mesoporous Silica Nanoparticle Interaction with Hairy Root Cultures during Nanoharvesting of Biomolecules. Adv Biol (Weinh) 2021; 5:e2000173. [PMID: 33729698 DOI: 10.1002/adbi.202000173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 01/11/2021] [Indexed: 11/06/2022]
Abstract
Cellular uptake and expulsion mechanisms of engineered mesoporous silica nanoparticles (MSNPs) are important in their design for novel biomolecule isolation and delivery applications such as nanoharvesting, defined as using nanocarriers to transport and isolate valuable therapeutics (secondary metabolites) out of living plant organ cultures (e.g., hairy roots). Here, temperature-dependent MSNP uptake and recovery processes in hairy roots are examined as a function of surface chemistry. MSNP uptake into hairy roots and time-dependent expulsion are quantified using Ti content (present for biomolecule binding) and fluorescence spectroscopy of fluorescently tagged MSNPs, respectively. The results suggest that functionalization and surface charge (regulated by amine group attachment) play the biggest role in the effectiveness of uptake and recovery. Comparison of MSNP interactions with hairy roots at 4 and 23 °C shows that weakly charged MSNPs functionalized only with Ti are taken up and expelled by thermally activated mechanisms, while amine-modified positively charged particles are taken up and expelled mainly by direct penetration of cell walls. Amine-functionalized MSNPs move spontaneously in and out of plant cells by dynamic exchange with a residence time of 20 ± 5 min, suggesting promise as a biomolecule nanoharvesting platform for plant organ cultures.
Collapse
Affiliation(s)
- Md Arif Khan
- Department of Chemical and Materials Engineering, University of Kentucky, 177 F. Paul Anderson Tower, Lexington, KY, 40506, USA
| | - Madeleine Fugate
- Department of Chemical and Materials Engineering, University of Kentucky, 177 F. Paul Anderson Tower, Lexington, KY, 40506, USA
| | | | | | | | - Stephen E Rankin
- Department of Chemical and Materials Engineering, University of Kentucky, 177 F. Paul Anderson Tower, Lexington, KY, 40506, USA
| | - Barbara L Knutson
- Department of Chemical and Materials Engineering, University of Kentucky, 177 F. Paul Anderson Tower, Lexington, KY, 40506, USA
| |
Collapse
|
48
|
Endolysosomal TRPMLs in Cancer. Biomolecules 2021; 11:biom11010065. [PMID: 33419007 PMCID: PMC7825278 DOI: 10.3390/biom11010065] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 12/31/2020] [Accepted: 12/31/2020] [Indexed: 12/13/2022] Open
Abstract
Lysosomes, the degradative endpoints and sophisticated cellular signaling hubs, are emerging as intracellular Ca2+ stores that govern multiple cellular processes. Dys-homeostasis of lysosomal Ca2+ is intimately associated with a variety of human diseases including cancer. Recent studies have suggested that the Ca2+-permeable channels Transient Receptor Potential (TRP) Mucolipins (TRPMLs, TRPML1-3) integrate multiple processes of cell growth, division and metabolism. Dysregulation of TRPMLs activity has been implicated in cancer development. In this review, we provide a summary of the latest development of TRPMLs in cancer. The expression of TRPMLs in cancer, TRPMLs in cancer cell nutrient sensing, TRPMLs-mediated lysosomal exocytosis in cancer development, TRPMLs in TFEB-mediated gene transcription of cancer cells, TRPMLs in bacteria-related cancer development and TRPMLs-regulated antitumor immunity are discussed. We hope to guide readers toward a more in-depth discussion of the importance of lysosomal TRPMLs in cancer progression and other human diseases.
Collapse
|
49
|
Wang H, Zhang Z, Guan J, Lu W, Zhan C. Unraveling GLUT-mediated transcytosis pathway of glycosylated nanodisks. Asian J Pharm Sci 2021; 16:120-128. [PMID: 33613735 PMCID: PMC7878461 DOI: 10.1016/j.ajps.2020.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/08/2020] [Accepted: 07/06/2020] [Indexed: 12/14/2022] Open
Abstract
Glucose transporter (GLUT)-mediated transcytosis has been validated as an efficient method to cross the blood-brain barrier and enhance brain transport of nanomedicines. However, the transcytosis process remains elusive. Glycopeptide-modified nanodisks (Gly-A7R-NDs), which demonstrated high capacity of brain targeting via GLUT-mediated transcytosis in our previous reports, were utilized to better understand the whole transcytosis process. Gly-A7R-NDs internalized brain capillary endothelial cells mainly via GLUT-mediated/clathrin dependent endocytosis and macropinocytosis. The intracellular Gly-A7R-NDs remained intact, and the main excretion route of Gly-A7R-NDs was lysosomal exocytosis. Glycosylation of nanomedicine was crucial in GLUT-mediated transcytosis, while morphology did not affect the efficiency. This study highlights the pivotal roles of lysosomal exocytosis in the process of GLUT-mediated transcytosis, providing a new impetus to development of brain targeting drug delivery by accelerating lysosomal exocytosis.
Collapse
Affiliation(s)
- Huan Wang
- Department of Pharmacology, School of Basic Medical Sciences and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
- Center of Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
- School of Pharmacy, Fudan University and Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Zui Zhang
- Department of Pharmacology, School of Basic Medical Sciences and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
| | - Juan Guan
- Department of Pharmacology, School of Basic Medical Sciences and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
- School of Pharmacy, Fudan University and Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Weiyue Lu
- School of Pharmacy, Fudan University and Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Changyou Zhan
- Department of Pharmacology, School of Basic Medical Sciences and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, China
- Center of Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
- School of Pharmacy, Fudan University and Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| |
Collapse
|
50
|
Qi Z, Jiang C, Gao H, Wang Y, Zhang Q, Zhang W, Liu J. Endocytic recycling as cellular trafficking fate of simvastatin-loaded discoidal reconstituted high-density lipoprotein to coordinate cholesterol efflux and drug influx. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 32:102323. [PMID: 33186693 DOI: 10.1016/j.nano.2020.102323] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 10/14/2020] [Accepted: 10/20/2020] [Indexed: 11/29/2022]
Abstract
Reconstituted high-density lipoproteins (rHDLs) hold promise as nanocarriers for atherosclerosis-targeted delivery, with biofunctions typified by mediating cholesterol efflux. The paradox is how rHDL offloads the delivered drugs into atherosclerotic foam cells, while simultaneously transferring cholesterol out of cells. Herein, simvastatin-loaded discoidal rHDL (ST-d-rHDL), constructed based on established paradigms, was employed to investigate its basic trafficking mechanism in foam cells. As proved, ST-d-rHDL was resecreted via lysosomal and Golgi apparatus-recycling endosome-mediated pathways following clathrin-mediated endocytosis. And the resecretion ratio reached 60% within 6-h chase with excessive ST-d-rHDLs. During the rHDL resecretion, 39% of cellular cholesterol efflux was detected, accompanied by 85% of the encapsulated cargo released intracellularly. Furthermore, the recycling rate was demonstrated to be promoted by smaller rHDL size and higher cellular lipid contents. Collectively, endocytic recycling confers the synergism in ST-d-rHDL to coordinate cholesterol efflux and intracellular drug release, providing new insights into design of biofunctional rHDL.
Collapse
Affiliation(s)
- Zitong Qi
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, PR China; State Key Laboratory of Long-Acting and Targeting Drug Delivery System, Shandong Luye Pharmaceutical Co., Ltd, Yantai, PR China
| | - Cuiping Jiang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, PR China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, PR China
| | - Hai Gao
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, PR China
| | - Yanyan Wang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, PR China
| | - Qiqi Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, PR China
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, PR China.
| | - Jianping Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, PR China.
| |
Collapse
|