1
|
Cao KY, Zhang D, Bai LB, Yan TM, Chen Y, Jiang YY, Jiang ZH. Targeting NUCKS1 with a fragment of tRNA Asn(GUU) of Chinese yew for the treatment of colorectal cancer. Noncoding RNA Res 2025; 11:38-47. [PMID: 39736854 PMCID: PMC11683283 DOI: 10.1016/j.ncrna.2024.11.002] [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: 08/24/2024] [Revised: 10/22/2024] [Accepted: 11/11/2024] [Indexed: 01/01/2025] Open
Abstract
Despite the discovery of numerous oncogenes in colorectal cancer (CRC), the development of associated drugs is limited, posing a significant challenge for CRC treatment. Identification of novel druggable targets is therefore crucial for the therapeutic development of CRC. Here, we report the first investigation on therapeutics targeting the potent oncogene NUCKS1 to suppress cancer progression. NUCKS1-orientated bioinformatics screening of NUCKS1 inhibitors from our library of tRNA fragments originated from medicinal plants identified tRF-T36, a 5' tRNA fragment of tRNAAsn(GUU) of Chinese yew (Taxus chinensis), exhibiting stronger inhibitory effects than taxol against CRC progression. Mechanistically, tRF-T36 binds directly to the 3' UTR of NUCKS1 mRNA to downregulate its expressions via RNAi pathway. High-throughput RNA sequencing indicated that the downregulated NUCKS1 induced by tRF-T36 further inhibits PI3K/Akt pathway, as verified by the significantly efficacy decrease of tRF-T36 mimic in co-treatment with 740Y-P, an agonist of PI3K/Akt pathway. Collectively, our findings emphasize the importance of NUCKS1 as a promising druggable target for CRC. Furthermore, the present study provides the first siRNA sequence, tRF-T36 mimic, as small RNA drug candidate, thereby shedding light on CRC therapeutics.
Collapse
Affiliation(s)
- Kai-Yue Cao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau SAR, China
| | - Da Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau SAR, China
| | - Long-Bo Bai
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau SAR, China
| | - Tong-Meng Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau SAR, China
| | - Yan Chen
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
| | - Yu-Yang Jiang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, Guangdong, China
| | - Zhi-Hong Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau SAR, China
| |
Collapse
|
2
|
Arya BD, Mittal S, Joshi P, Pandey AK, Ramirez-Vick JE, Gupta G, Singh SP. Graphene oxide-chloroquine conjugate induces DNA damage in A549 lung cancer cells through autophagy modulation. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2025; 16:316-332. [PMID: 40041432 PMCID: PMC11878127 DOI: 10.3762/bjnano.16.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 01/28/2025] [Indexed: 03/06/2025]
Abstract
Autophagy is a highly regulated catabolic process by which unnecessary, dysfunctional, or damaged proteins and other cellular components are degraded and recycled to promote cellular differentiation, survival, and development. In response to endogenous or exogenous stresses, cancer cells use autophagy pathways for survival through activation of complex DNA damage repair (DDR) mechanisms. In the present study, we demonstrated the genotoxicity induced in A549 lung cancer cells by exposure to the GO-Chl nanoconjugate and elucidated the role of autophagy modulation in harnessing the DNA-damage response. GO-Chl causes loss of plasma membrane integrity, cell cycle arrest, and significant genotoxicity in A549 cells. Further, elevated expression of key autophagy proteins beclin-1, ATG-7, LC-3-I/II, and SQSTM1/p62 reveal that inhibition of autophagy plays a crucial role in regulating DDR capabilities of cancer cells. The results indicate that the interplay between DDR and autophagy pathways may open new paradigms for developing effective combinatorial nanoscale drug systems against multidrug-resistance cancers.
Collapse
Affiliation(s)
- Braham Dutt Arya
- CSIR-National Physical Laboratory, Dr K. S. Krishanan Marg, New Delhi-12, India
- Academy of Scientific & Innovative Research (AcSIR), New Delhi-20, India
- Department of Higher Education, Shiksha Sadan, Sector-5, Panchkula-134114, India
| | - Sandeep Mittal
- Academy of Scientific & Innovative Research (AcSIR), New Delhi-20, India
- CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 31, Mahatma Gandhi Marg, Lucknow-226001, India
| | - Prachi Joshi
- CSIR-National Physical Laboratory, Dr K. S. Krishanan Marg, New Delhi-12, India
| | - Alok Kumar Pandey
- Academy of Scientific & Innovative Research (AcSIR), New Delhi-20, India
- CSIR-Indian Institute of Toxicology Research (CSIR-IITR), 31, Mahatma Gandhi Marg, Lucknow-226001, India
| | - Jaime E Ramirez-Vick
- Department of Biomedical, Industrial & Human Factors Engineering, Wright State University, Dayton, Ohio 45435, United States
| | - Govind Gupta
- CSIR-National Physical Laboratory, Dr K. S. Krishanan Marg, New Delhi-12, India
- Academy of Scientific & Innovative Research (AcSIR), New Delhi-20, India
| | - Surinder P Singh
- CSIR-National Physical Laboratory, Dr K. S. Krishanan Marg, New Delhi-12, India
- Academy of Scientific & Innovative Research (AcSIR), New Delhi-20, India
| |
Collapse
|
3
|
Clark C, Barzegar Behrooz A, Cordani M, Shojaei S, Ghavami S. Assessing Autophagy Flux in Glioblastoma Temozolomide Resistant Cells. Methods Mol Biol 2025; 2879:225-238. [PMID: 39331341 DOI: 10.1007/7651_2024_571] [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: 09/28/2024]
Abstract
Autophagy is a critical cellular process involved in the degradation and recycling of cytoplasmic components, playing a dual role in cancer by either promoting cell survival or facilitating cell death. In glioblastoma (GB), autophagy has been implicated in resistance to the chemotherapeutic agent temozolomide (TMZ). This study presents a novel method to accurately measure autophagy flux in TMZ-resistant glioblastoma cells, combining advanced imaging techniques with biochemical assays. By quantifying key autophagy markers such as LC3-II and SQSTM1, our approach provides detailed insights into the dynamic processes of autophagosome formation and clearance under therapeutic stress. This method advances our understanding of autophagy in GB chemoresistance and has significant implications for the development of autophagy-targeted therapies. The ability to monitor and manipulate autophagy flux in real time offers a promising avenue for monitoring and understanding TMZ resistance and improving patient outcomes in glioblastoma treatment.
Collapse
Affiliation(s)
- Courtney Clark
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University of Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Shahla Shojaei
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
- Faculty of Medicine in Zabrze, University of Technology in Katowice, Zabrze, Poland.
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
4
|
Flasz B, Babczyńska A, Tarnawska M, Ajay AK, Kędziorski A, Napora-Rutkowski Ł, Augustyniak M. Graphene oxide in low concentrations can change mitochondrial potential, autophagy, and apoptosis paths in two strains of invertebrates with different life strategies. Biochem Biophys Res Commun 2024; 736:150898. [PMID: 39467354 DOI: 10.1016/j.bbrc.2024.150898] [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: 09/17/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 10/30/2024]
Abstract
Nanoparticles, like graphene oxide (GO), are particles with unique physiochemical properties that enable their wide application in various areas of life. The effects of GO on individual cell organelles like mitochondria and the effects of interactions are worth investigating, as they can activate multiple cellular processes, such as autophagy or apoptosis. Mitochondrial injury plays an essential role in the majority of cell death routines. In the project, we investigated cell health status measured as mitochondrial inner membrane depolarization, autophagy, and apoptosis induction during long-term GO administration in food (0.02 μg g-1 and 0.2 μg g-1 of food). Two unique Acheta domesticus strains that differ in life strategy were used: wild-type and long-lived at three different life stages (larva, young adult, mature adult). The changes in mitochondrial trans-membrane potential were marked in the wild-type strain. The autophagy was lower in all GO-treated groups in both strains, and the apoptosis was lower in both strains in the mature adult crickets. Low GO concentrations treatment for the whole life, despite mitochondrial dysfunction, may lead to inhibition of autophagy and apoptosis by arresting the cell cycle for the duration of repair, and other repair tools are involved in the process of restoring homeostasis.
Collapse
Affiliation(s)
- Barbara Flasz
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Poland.
| | - Agnieszka Babczyńska
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Poland
| | - Monika Tarnawska
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Poland
| | - Amrendra K Ajay
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Andrzej Kędziorski
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Poland
| | - Łukasz Napora-Rutkowski
- Polish Academy of Sciences, Institute of Ichthyobiology and Aquaculture in Gołysz, 43-520, Chybie, Poland
| | - Maria Augustyniak
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Poland
| |
Collapse
|
5
|
Siquan L, Weilin C, Xiuwen C, Meiyan Z, Weihong G, Xiaoli F. Evaluating the safety and efficiency of nanomaterials: A focus on mitochondrial health. Biomed Pharmacother 2024; 180:117484. [PMID: 39316969 DOI: 10.1016/j.biopha.2024.117484] [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: 07/23/2024] [Revised: 09/09/2024] [Accepted: 09/20/2024] [Indexed: 09/26/2024] Open
Abstract
Nanomaterials (NMs) have extensive application potential in drug delivery, tissue engineering, and various other domains, attributable to their exceptional physical and chemical properties. Nevertheless, an increasing body of literature underscores the diverse safety risks are associated with NMs upon interaction with the human body, including oxidative stress and programmed cell death. Mitochondria, serving as cellular energy factories, play a pivotal role in energy metabolism and the regulation of cell fate. Organs with substantial energy demands, including the heart and brain, are highly sensitive to mitochondrial integrity, with mitochondrial impairment potentially resulting in significant dysfunction and pathologies such as as heart failure and neurodegenerative disease. This review elucidates the pathways by which NMs translocate into mitochondria, their intracellular dynamics, and their impact on mitochondrial morphology, respiratory chain activity, and metabolic processes. We further investigate associated molecular mechanisms, including mitochondrial dynamic imbalance, calcium overload, and oxidative stress, and elucidate the pivotal roles of mitochondria in different forms of programmed cell death such as apoptosis and autophagy. Finally, we offer recommendations regarding the safety and efficacy of NMs for medical applications. By systematically analyzing the interactions and molecular mechanisms between NMs and mitochondria, this paper aims to enhance the toxicological evaluation framework of NMs and provide a foundational reference and theoretical basis for their clinical utilization.
Collapse
Affiliation(s)
- Liu Siquan
- Stomatology Hospital, School of Stomatology, Southern Medical University, Guangzhou 510515, China
| | - Cheng Weilin
- Stomatology Hospital, School of Stomatology, Southern Medical University, Guangzhou 510515, China
| | - Chen Xiuwen
- Stomatology Hospital, School of Stomatology, Southern Medical University, Guangzhou 510515, China
| | - Zou Meiyan
- Stomatology Hospital, School of Stomatology, Southern Medical University, Guangzhou 510515, China
| | - Guo Weihong
- Department of General Surgery, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China.
| | - Feng Xiaoli
- Stomatology Hospital, School of Stomatology, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
6
|
Deng S, Zhao Q, Liu D, Xiong Z, Zhang S, Zhang X, Wu F, Xing B. Black phosphorus nanosheets induce autophagy dysfunction by a size- and surface modification-related impairment of lysosomes in macrophages. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117073. [PMID: 39332199 DOI: 10.1016/j.ecoenv.2024.117073] [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: 05/06/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 09/29/2024]
Abstract
The widespread application of black phosphorus nanosheets (BPNSs) raises concerns about their potential impact on human health. Although that the autophagy-inducing properties of BPNSs in cancer cells are documented, their effects on macrophages-key components of the immune system and the mechanisms involved remain obscure, especially in terms of the influences of BPNS the size and surface modifications on the autophagic process. This study investigated the effects of bare BPNSs and PEGylated BPNSs (BP-PEG) on macrophage autophagy and its underlying mechanisms by comprehensive biochemical analyses. The results indicated that both BPNSs and BP-PEG are internalized by RAW264.7 cells through phagocytosis and caveolin-dependent endocytosis, leading to lysosomal accumulation. The internalized BPNSs induced mitochondrial dysfunction, which subsequently elevated the NAD+/NADH ratio and activated the SIRT-1 pathway, initiating autophagy. However, BPNSs disrupted the autophagic flux by impairing autolysosome formation, leading to apoptosis in a size-dependent manner. In contrast, BP-PEG preserved lysosomal integrity, maintaining autophagic activity and cell viability. These findings deepen our understanding of the influence of nanosheet size and surface modifications on macrophage autophagy, contributing to the formulation of regulatory guidelines to minimize the potential adverse effects and health risks associated with BPNS utilization in various applications.
Collapse
Affiliation(s)
- Shuo Deng
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China; National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Institute of Eco-environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qing Zhao
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Institute of Eco-environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, China
| | - Daxu Liu
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiqiang Xiong
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Institute of Eco-environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, China
| | - Siyu Zhang
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China; National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Institute of Eco-environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, China
| | - Xuejiao Zhang
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China; National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Institute of Eco-environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, China.
| | - Fengchang Wu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Baoshan Xing
- Stockbridge School of Agriculture, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
7
|
Yan X, An N, Zhang Z, Qiu Q, Yang D, Wei P, Zhang X, Qiu L, Guo J. Graphene Oxide Quantum Dots-Preactivated Dental Pulp Stem Cells/GelMA Facilitates Mitophagy-Regulated Bone Regeneration. Int J Nanomedicine 2024; 19:10107-10128. [PMID: 39381026 PMCID: PMC11460356 DOI: 10.2147/ijn.s480979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/02/2024] [Indexed: 10/10/2024] Open
Abstract
Background In bone tissue engineering (BTE), cell-laden scaffolds offer a promising strategy for repairing bone defects, particularly when host cell regeneration is insufficient due to age or disease. Exogenous stem cell-based BTE requires bioactive factors to activate these cells. Graphene oxide quantum dots (GOQDs), zero-dimensional derivatives of graphene oxide, have emerged as potential osteogenic nanomedicines. However, constructing biological scaffolds with GOQDs and elucidating their biological mechanisms remain critical challenges. Methods We utilized GOQDs with a particle size of 10 nm, characterized by a surface rich in C-O-H and C-O-C functional groups. We developed a gelatin methacryloyl (GelMA) hydrogel incorporated with GOQDs-treated dental pulp stem cells (DPSCs). These constructs were transplanted into rat calvarial bone defects to estimate the effectiveness of GOQDs-induced DPSCs in repairing bone defects while also investigating the molecular mechanism underlying GOQDs-induced osteogenesis in DPSCs. Results GOQDs at 5 μg/mL significantly enhanced the osteogenic differentiation of DPSCs without toxicity. The GOQDs-induced DPSCs showed active osteogenic potential in three-dimensional cell culture system. In vivo, transplantation of GOQDs-preactivated DPSCs/GelMA composite effectively facilitated calvarial bone regeneration. Mechanistically, GOQDs stimulated mitophagy flux through the phosphatase-and-tensin homolog-induced putative kinase 1 (PINK1)/Parkin E3 ubiquitin ligase (PRKN) pathway. Notably, inhibiting mitophagy with cyclosporin A prevented the osteogenic activity of GOQDs. Conclusion This research presents a well-designed bionic GOQDs/DPSCs/GelMA composite scaffold and demonstrated its ability to promote bone regeneration by enhancing mitophagy. These findings highlight the significant potential of this composite for application in BTE and underscore the crucial role of mitophagy in promoting the osteogenic differentiation of GOQDs-induced stem cells.
Collapse
Affiliation(s)
- Xiaoyuan Yan
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Na An
- Department of Orthodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Zeying Zhang
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Qiujing Qiu
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Di Yang
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Penggong Wei
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Xiyue Zhang
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Lihong Qiu
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| | - Jiajie Guo
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, People’s Republic of China
| |
Collapse
|
8
|
Park J, Wu Y, Suk Kim J, Byun J, Lee J, Oh YK. Cytoskeleton-modulating nanomaterials and their therapeutic potentials. Adv Drug Deliv Rev 2024; 211:115362. [PMID: 38906478 DOI: 10.1016/j.addr.2024.115362] [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: 03/14/2024] [Revised: 05/25/2024] [Accepted: 06/16/2024] [Indexed: 06/23/2024]
Abstract
The cytoskeleton, an intricate network of protein fibers within cells, plays a pivotal role in maintaining cell shape, enabling movement, and facilitating intracellular transport. Its involvement in various pathological states, ranging from cancer proliferation and metastasis to the progression of neurodegenerative disorders, underscores its potential as a target for therapeutic intervention. The exploration of nanotechnology in this realm, particularly the use of nanomaterials for cytoskeletal modulation, represents a cutting-edge approach with the promise of novel treatments. Inorganic nanomaterials, including those derived from gold, metal oxides, carbon, and black phosphorus, alongside organic variants such as peptides and proteins, are at the forefront of this research. These materials offer diverse mechanisms of action, either by directly interacting with cytoskeletal components or by influencing cellular signaling pathways that, in turn, modulate the cytoskeleton. Recent advancements have introduced magnetic field-responsive and light-responsive nanomaterials, which allow for targeted and controlled manipulation of the cytoskeleton. Such precision is crucial in minimizing off-target effects and enhancing therapeutic efficacy. This review explores the importance of research into cytoskeleton-targeting nanomaterials for developing therapeutic interventions for a range of diseases. It also addresses the progress made in this field, the challenges encountered, and future directions for using nanomaterials to modulate the cytoskeleton. The continued exploration of nanomaterials for cytoskeleton modulation holds great promise for advancing therapeutic strategies against a broad spectrum of diseases, marking a significant step forward in the intersection of nanotechnology and medicine.
Collapse
Affiliation(s)
- Jinwon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yina Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jung Suk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Junho Byun
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| | - Jaiwoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
9
|
Zhang M, Song X, Liu S, Zhang N, Yang M, Gao P, Geng Z, Zuo L, Zhang X, Wang L, Wang Y, Li J, Hu J. Magnolin inhibits intestinal epithelial cell apoptosis alleviating Crohn's disease-like colitis by suppressing the PI3K/AKT signalling pathway. Int Immunopharmacol 2024; 134:112181. [PMID: 38733829 DOI: 10.1016/j.intimp.2024.112181] [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: 03/25/2024] [Revised: 04/26/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND AND AIMS Previous reports have shown that preventing excessive intestinal epithelial cell (IEC) apoptosis is a crucial approach for protecting the intestinal barrier in patients with Crohn's disease (CD). Magnolin (MGL) has various biological activities, including antiapoptotic activities, but its role in CD has largely not been determined. This study investigated how MGL impacts CD-like colitis and the underlying mechanism involved. METHODS Mice were treated with TNBS to establish a disease model, and these mice were used to assess the therapeutic effects of MGL on CD-like colitis. TNF-α-treated colon organoids were used to evaluate the impact of MGL on intestinal barrier function and IEC apoptosis. Enrichment analysis was performed to examine the potential pathways through which MGL inhibits IEC apoptosis. Finally, rescue experiments showed the mechanism by which MGL suppresses IEC apoptosis. RESULTS The animal experiments demonstrated that MGL treatment alleviated the weight loss, colon shortening, elevated disease activity index (DAI) scores, increased colitis histological scores and upregulated inflammatory factor expression that were observed in model mice. MGL ameliorated intestinal barrier dysfunction and the loss of tight junction (TJ) proteins (ZO-1 and Claudin-1) by inhibiting IEC apoptosis in both TNBS-treated mice and TNF-α-treated colon organoids. MGL inhibited the PI3K/AKT signalling pathway, thus safeguarding the intestinal barrier and alleviating CD-like colitis in vivo and in vitro. CONCLUSIONS MGL improves the intestinal barrier integrity and prevents CD-like colitis by inhibiting IEC apoptosis. The potential mechanism of its anti-apoptotic impact on IECs could be associated with the PI3K/AKT pathway, presenting novel approaches and avenues for the clinical management of CD.
Collapse
Affiliation(s)
- Min Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China; Department of Laboratory Medicine, Bengbu Medical University, Bengbu, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
| | - Xue Song
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
| | - Shengbao Liu
- Department of Pathology, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Nuo Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China; Department of Laboratory Medicine, Bengbu Medical University, Bengbu, China
| | - Ming Yang
- Department of Laboratory Medicine, Bengbu Medical University, Bengbu, China
| | - Pengcheng Gao
- Department of Laboratory Medicine, Bengbu Medical University, Bengbu, China
| | - Zhijun Geng
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
| | - Lugen Zuo
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China; Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Xiaofeng Zhang
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
| | - Lian Wang
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China; Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Yueyue Wang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
| | - Jing Li
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
| | - Jianguo Hu
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China.
| |
Collapse
|
10
|
Peng J, Li S, Ti H. Sensitize Tumor Immunotherapy: Immunogenic Cell Death Inducing Nanosystems. Int J Nanomedicine 2024; 19:5895-5930. [PMID: 38895146 PMCID: PMC11184231 DOI: 10.2147/ijn.s457782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Low immunogenicity of tumors poses a challenge in the development of effective tumor immunotherapy. However, emerging evidence suggests that certain therapeutic approaches, such as chemotherapy, radiotherapy, and phototherapy, can induce varying degrees of immunogenic cell death (ICD). This ICD phenomenon leads to the release of tumor antigens and the maturation of dendritic cells (DCs), thereby enhancing tumor immunogenicity and promoting immune responses. However, the use of a single conventional ICD inducer often fails to achieve in situ tumor ablation and establish long-term anti-tumor immune responses. Furthermore, the induction of ICD induction varies among different approaches, and the distribution of the therapeutic agent within the body influences the level of ICD and the occurrence of toxic side effects. To address these challenges and further boost tumor immunity, researchers have explored nanosystems as inducers of ICD in combination with tumor immunotherapy. This review examines the mechanisms of ICD and different induction methods, with a specific focus on the relationship between ICD and tumor immunity. The aim is to explore the research advancements utilizing various nanomaterials to enhance the body's anti-tumor effects by inducing ICD. This paper aims to contribute to the development and clinical application of nanomaterial-based ICD inducers in the field of cancer immunotherapy by providing important theoretical guidance and practical references.
Collapse
Affiliation(s)
- Jianlan Peng
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Shiying Li
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Huihui Ti
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
- Guangdong Province Precise Medicine and Big Data Engineering Technology Research Center for Traditional Chinese Medicine, Guangzhou, People’s Republic of China
| |
Collapse
|
11
|
Lai X, Wang M, Zhang Z, Chen S, Tan X, Liu W, Liang H, Li L, Shao L. ZNPs reduce epidermal mechanical strain resistance by promoting desmosomal cadherin endocytosis via mTORC1-TFEB-BLOC1S3 axis. J Nanobiotechnology 2024; 22:312. [PMID: 38840221 PMCID: PMC11151536 DOI: 10.1186/s12951-024-02519-z] [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: 01/09/2024] [Accepted: 05/01/2024] [Indexed: 06/07/2024] Open
Abstract
Zinc oxide nanoparticles (ZNPs) are widely used in sunscreens and nanomedicines, and it was recently confirmed that ZNPs can penetrate stratum corneum into deep epidermis. Therefore, it is necessary to determine the impact of ZNPs on epidermis. In this study, ZNPs were applied to mouse skin at a relatively low concentration for one week. As a result, desmosomes in epidermal tissues were depolymerized, epidermal mechanical strain resistance was reduced, and the levels of desmosomal cadherins were decreased in cell membrane lysates and increased in cytoplasmic lysates. This finding suggested that ZNPs promote desmosomal cadherin endocytosis, which causes desmosome depolymerization. In further studies, ZNPs were proved to decrease mammalian target of rapamycin complex 1 (mTORC1) activity, activate transcription factor EB (TFEB), upregulate biogenesis of lysosome-related organelle complex 1 subunit 3 (BLOC1S3) and consequently promote desmosomal cadherin endocytosis. In addition, the key role of mTORC1 in ZNP-induced decrease in mechanical strain resistance was determined both in vitro and in vivo. It can be concluded that ZNPs reduce epidermal mechanical strain resistance by promoting desmosomal cadherin endocytosis via the mTORC1-TFEB-BLOC1S3 axis. This study helps elucidate the biological effects of ZNPs and suggests that ZNPs increase the risk of epidermal fragmentation.
Collapse
Affiliation(s)
- Xuan Lai
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510515, China
| | - Menglei Wang
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhen Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510515, China
| | - Suya Chen
- Hospital of Stomatology, Guanghua school of Stomatology, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiner Tan
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510515, China
| | - Wenjing Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510515, China
| | - Huimin Liang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510515, China
| | - Li Li
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Longquan Shao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
12
|
Song Q, Song C, Chen X, Xiong Y, He Z, Su X, Zhou J, Ke H, Dong C, Liao W, Yang S. Oxalate regulates crystal-cell adhesion and macrophage metabolism via JPT2/PI3K/AKT signaling to promote the progression of kidney stones. J Pharm Anal 2024; 14:100956. [PMID: 39035219 PMCID: PMC11259813 DOI: 10.1016/j.jpha.2024.02.010] [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: 09/25/2023] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 07/23/2024] Open
Abstract
Oxalate is an organic dicarboxylic acid that is a common component of plant foods. The kidneys are essential organs for oxalate excretion, but excessive oxalates may induce kidney stones. Jupiter microtubule associated homolog 2 (JPT2) is a critical molecule in Ca2+ mobilization, and its intrinsic mechanism in oxalate exposure and kidney stones remains unclear. This study aimed to reveal the mechanism of JPT2 in oxalate exposure and kidney stones. Genetic approaches were used to control JPT2 expression in cells and mice, and the JPT2 mechanism of action was analyzed using transcriptomics and untargeted metabolomics. The results showed that oxalate exposure triggered the upregulation of JPT2, which is involved in nicotinic acid adenine dinucleotide phosphate (NAADP)-mediated Ca2+ mobilization. Transcriptomic analysis revealed that cell adhesion and macrophage inflammatory polarization were inhibited by JPT2 knockdown, and these were dominated by phosphatidylinositol 3-kinase (PI3K)/AKT signaling, respectively. Untargeted metabolomics indicated that JPT2 knockdown inhibited the production of succinic acid semialdehyde (SSA) in macrophages. Furthermore, JPT2 deficiency in mice inhibited kidney stones mineralization. In conclusion, this study demonstrates that oxalate exposure facilitates kidney stones by promoting crystal-cell adhesion, and modulating macrophage metabolism and inflammatory polarization via JPT2/PI3K/AKT signaling.
Collapse
Affiliation(s)
- Qianlin Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Chao Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xin Chen
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yunhe Xiong
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ziqi He
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaozhe Su
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jiawei Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hu Ke
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Caitao Dong
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wenbiao Liao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Sixing Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| |
Collapse
|
13
|
Liu J, Du C, Chen H, Huang W, Lei Y. Nano-Micron Combined Hydrogel Microspheres: Novel Answer for Minimal Invasive Biomedical Applications. Macromol Rapid Commun 2024; 45:e2300670. [PMID: 38400695 DOI: 10.1002/marc.202300670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/05/2024] [Indexed: 02/25/2024]
Abstract
Hydrogels, key in biomedical research for their hydrophilicity and versatility, have evolved with hydrogel microspheres (HMs) of micron-scale dimensions, enhancing their role in minimally invasive therapeutic delivery, tissue repair, and regeneration. The recent emergence of nanomaterials has ushered in a revolutionary transformation in the biomedical field, which demonstrates tremendous potential in targeted therapies, biological imaging, and disease diagnostics. Consequently, the integration of advanced nanotechnology promises to trigger a new revolution in the realm of hydrogels. HMs loaded with nanomaterials combine the advantages of both hydrogels and nanomaterials, which enables multifaceted functionalities such as efficient drug delivery, sustained release, targeted therapy, biological lubrication, biochemical detection, medical imaging, biosensing monitoring, and micro-robotics. Here, this review comprehensively expounds upon commonly used nanomaterials and their classifications. Then, it provides comprehensive insights into the raw materials and preparation methods of HMs. Besides, the common strategies employed to achieve nano-micron combinations are summarized, and the latest applications of these advanced nano-micron combined HMs in the biomedical field are elucidated. Finally, valuable insights into the future design and development of nano-micron combined HMs are provided.
Collapse
Affiliation(s)
- Jiacheng Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chengcheng Du
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hong Chen
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wei Huang
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yiting Lei
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
14
|
Jiang Y, Zhou D, Jiang Y. Three-dimensional bioprinted GelMA/GO composite hydrogel for stem cell osteogenic differentiation both in vitro and in vivo. J Biomater Appl 2024; 38:1087-1099. [PMID: 38561006 DOI: 10.1177/08853282241243337] [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: 04/04/2024]
Abstract
In this study, we evaluated the use of graphene oxide (GO) mixed with methyl methacrylate gelatin (GelMA) for the construction of a microenvironmental implant to repair bone defects in orthopedic surgery. A scaffold containing a GelMA/GO composite with mesenchymal stem cells (MSCs) was constructed using three-dimensional bioprinting. The survival and osteogenic capacity of MSCs in the composite bioink were evaluated using cell viability and proliferation assays, osteogenesis-related gene expression analysis, and implantation under the skin of nude mice. The printing process had little effect on cell viability. We found that GO enhanced cell proliferation but had no significant effect on cell viability. In vitro experiments suggested that GO promoted material-cell interactions and the expression of osteogenesis-related genes. In vivo experiments showed that GO decreased the degradation time of the material and increased calcium nodule deposition. In contrast to pure GelMA, the addition of GO created a suitable microenvironment to promote the differentiation of loaded exogenous MSCs in vitro and in vivo, providing a basis for the repair of bone defects.
Collapse
Affiliation(s)
- Yerong Jiang
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Dezhi Zhou
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing, People's Republic of China
| | - Yanan Jiang
- Department of Burns and Plastic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
15
|
Castagnola V, Tomati V, Boselli L, Braccia C, Decherchi S, Pompa PP, Pedemonte N, Benfenati F, Armirotti A. Sources of biases in the in vitro testing of nanomaterials: the role of the biomolecular corona. NANOSCALE HORIZONS 2024; 9:799-816. [PMID: 38563642 DOI: 10.1039/d3nh00510k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The biological fate of nanomaterials (NMs) is driven by specific interactions through which biomolecules, naturally adhering onto their surface, engage with cell membrane receptors and intracellular organelles. The molecular composition of this layer, called the biomolecular corona (BMC), depends on both the physical-chemical features of the NMs and the biological media in which the NMs are dispersed and cells grow. In this work, we demonstrate that the widespread use of 10% fetal bovine serum in an in vitro assay cannot recapitulate the complexity of in vivo systemic administration, with NMs being transported by the blood. For this purpose, we undertook a comparative journey involving proteomics, lipidomics, high throughput multiparametric in vitro screening, and single molecular feature analysis to investigate the molecular details behind this in vivo/in vitro bias. Our work indirectly highlights the need to introduce novel, more physiological-like media closer in composition to human plasma to produce realistic in vitro screening data for NMs. We also aim to set the basis to reduce this in vitro-in vivo mismatch, which currently limits the formulation of NMs for clinical settings.
Collapse
Affiliation(s)
- Valentina Castagnola
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Valeria Tomati
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, Via Gaslini 5, 16147 Genova, Italy
| | - Luca Boselli
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Clarissa Braccia
- Analytical Chemistry Facility, Istituto Italiano di Tecnologia, Via Morego 30, Genova, 16163, Italy.
| | - Sergio Decherchi
- Data Science and Computation Facility, Istituto Italiano di Tecnologia, via Morego, 30, Genova, 16163, Italy
| | - Pier Paolo Pompa
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Nicoletta Pedemonte
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, Via Gaslini 5, 16147 Genova, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Largo Rosanna Benzi 10, 16132 Genova, Italy.
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Andrea Armirotti
- Analytical Chemistry Facility, Istituto Italiano di Tecnologia, Via Morego 30, Genova, 16163, Italy.
| |
Collapse
|
16
|
Liu J, Zhou W, Yang L, Li Y, Qiu J, Fu X, Ren P, Guo F, Zhou Y, Liu J, Chen P, DiSanto ME, Zhang X. STEAP4 modulates cell proliferation and oxidative stress in benign prostatic hyperplasia. Cell Signal 2024; 113:110933. [PMID: 37866665 DOI: 10.1016/j.cellsig.2023.110933] [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: 06/18/2023] [Revised: 09/08/2023] [Accepted: 10/18/2023] [Indexed: 10/24/2023]
Abstract
Benign prostatic hyperplasia (BPH) is a quite common chronic disease plagued elderly men and its etiology remains unclear. It was reported that the six-transmembrane epithelial antigen of prostate 4 (STEAP4) could modulate cell proliferation/apoptosis ratio and oxidative stress in cancers. Our current study aimed to explore the expression, biological function, and underlying mechanism of STEAP4 in BPH progress. Human prostate tissues and cell lines were utilized. qRT-PCR and immunofluorescence staining were employed. STEAP4 knockdown (STEAP4-KD) or STEAP4 overexpression (STEAP4-OE) cell models were established. Cell proliferation, cell cycle, apoptosis, and reactive oxygen species (ROS) were determined by cell counting kit-8 (CCK-8) assay and flow cytometry. Apoptosis-related proteins and antioxidant enzymes were identified by Western Blot. In addition, the epithelial-mesenchymal transition (EMT) process and fibrosis biomarker (collagen I and α-SMA) were analyzed. It was indicated that STEAP4 was mainly located in the prostate epithelium and upregulated in BPH tissues. STEAP4 deficiency induced apoptosis and inhibited cell survival, but had no effect on the cell cycle, fibrosis, and EMT process. In addition, ROS changes were observed in the STEAP4-KD model. Consistently, overproduction of STEAP4 suppressed apoptosis and promoted cell proliferation, as well as facilitated ROS production. We further examined AKT / mTOR, p38MAPK / p-p38MAPK, and WNT/ β-Catenin signaling pathway and demonstrated that STEAP4 regulated the proliferation and apoptosis of prostate cells through AKT / mTOR signaling, rather than p38MAPK / p-p38MAPK and WNT/ β-Catenin pathways. Furthermore, activating AKT / mTOR signaling with SC79 significantly reversed apoptosis triggered by STEAP4 deficiency, whereas suppressing AKT / mTOR signaling with MK2206 reduced the increase of cell viability triggered by STEAP4 overproduction. Our original data demonstrated that STEAP4 is crucial in the onset and progression of prostate hyperplasia and may become a new target for the treatment of BPH.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wei Zhou
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, China
| | - Liang Yang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yan Li
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jizhang Qiu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xun Fu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Pengfei Ren
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Feng Guo
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yongying Zhou
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jianmin Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ping Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Michael E DiSanto
- Department of Surgery and Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Xinhua Zhang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
17
|
Pang Y, Qu J, Zhang H, Cao Y, Ma X, Wang S, Wang J, Wu J, Zhang T. Nose-to-brain translocation and nervous system injury in response to indium tin oxide nanoparticles of long-term low-dose exposures. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 905:167314. [PMID: 37742979 DOI: 10.1016/j.scitotenv.2023.167314] [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] [Received: 05/19/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
Indium tin oxide (ITO) is a semiconductor nanomaterial with broad application in liquid crystal displays, solar cells, and electrochemical immune sensors. It is worth noting that, with the gradual increase in worker exposure opportunities, the exposure risk in occupational production cannot be ignored. At present, the toxicity of ITO mainly focuses on respiratory toxicity. ITO inhaled through the upper respiratory tract can cause pathological changes such as interstitial pneumonia and pulmonary fibrosis. Still, extrapulmonary toxicity after nanoscale ITO nanoparticle (ITO NPs) exposure, such as long-term effects on the central nervous system, should also be of concern. Therefore, we set up exposure dose experiments (0 mg·kg-1, 3.6 mg·kg-1, and 36 mg·kg-1) based on occupational exposure limits to treat C57BL/6 mice via nasal drops for 15 weeks. Moreover, we conducted a preliminary assessment of the neurotoxicity of ITO NPs (20-30 nm) in vivo. The results indicated that ITO NPs can cause diffuse inflammatory infiltrates in brain tissue, increased glial cell responsiveness, abnormal neuronal cell lineage transition, neuronal migration disorders, and neuronal apoptosis related to the oxidative stress induced by ITO NPs exposure. Hence, our findings provide useful information for the fuller risk assessment of ITO NPs after occupational exposure.
Collapse
Affiliation(s)
- Yanting Pang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Jing Qu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Haopeng Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Yuna Cao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Xinmo Ma
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Shile Wang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Jianli Wang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Jingying Wu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Ting Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| |
Collapse
|
18
|
Yang L, Liu YN, Gu Y, Guo Q. Deltonin enhances gastric carcinoma cell apoptosis and chemosensitivity to cisplatin via inhibiting PI3K/AKT/mTOR and MAPK signaling. World J Gastrointest Oncol 2023; 15:1739-1755. [PMID: 37969408 PMCID: PMC10631430 DOI: 10.4251/wjgo.v15.i10.1739] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/23/2023] [Accepted: 07/19/2023] [Indexed: 10/10/2023] Open
Abstract
BACKGROUND As an active ingredient derived from Dioscorea zingiberensis C.H. Wright, deltonin has been reported to show anti-cancer effects in a variety of malignancies. AIM To investigate the role and mechanism of action of deltonin in promoting gastric carcinoma (GC) cell apoptosis and chemosensitivity to cisplatin. METHODS The GC cell lines AGS, HGC-27, and MKN-45 were treated with deltonin and then subjected to flow cytometry and 3-(4,5-dimethylthiazol-2-yl)-3,5-diphenyltetrazolium bromide assays for cell apoptosis and viability determination. Western blot analysis was conducted to examine alterations in the expression of apoptosis-related proteins (Bax, Bid, Bad, and Fas), DNA repair-associated proteins (Rad51 and MDM2), and phosphatidylinositol 3-kinase/protein kinase B/mammalian target of the rapamycin (PI3K/AKT/mTOR) and p38-mitogen-activated protein kinase (MAPK) axis proteins. Additionally, the influence of deltonin on GC cell chemosensitivity to cisplatin was evaluated both in vitro and in vivo. RESULTS Deltonin treatment weakened viability, enhanced apoptosis, and dampened DNA repair in GC cell lines in a dose-dependent pattern. Furthermore, deltonin mitigated PI3K, AKT, mTOR, and p38-MAPK phosphorylation. HS-173, an inhibitor of PI3K, attenuated GC cell viability and abolished deltonin inhibition of GC cell viability and PI3K/AKT/mTOR and p38-MAPK pathway activation. Deltonin also promoted the chemosensitivity of GC cells to cisplatin via repressing GC cell proliferation and growth and accelerating apoptosis. CONCLUSION Deltonin can boost the chemosensitivity of GC cells to cisplatin via inactivating p38-MAPK and PI3K/AKT/mTOR signaling.
Collapse
Affiliation(s)
- Lin Yang
- Intensive Care Unit, Second Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Ya-Nan Liu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Yi Gu
- Nursing Department of Obstetrics and Gynecology, Second Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Qi Guo
- Department of Radiotherapy, Second Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| |
Collapse
|
19
|
Wang T, Yan L, Wang L, Sun J, Qu H, Ma Y, Song R, Tong X, Zhu J, Yuan Y, Gu J, Bian J, Liu Z, Zou H. VPS41-mediated incomplete autophagy aggravates cadmium-induced apoptosis in mouse hepatocytes. JOURNAL OF HAZARDOUS MATERIALS 2023; 459:132243. [PMID: 37562348 DOI: 10.1016/j.jhazmat.2023.132243] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 08/12/2023]
Abstract
Exposure to cadmium (Cd), an environmental heavy metal contaminant, is a serious threat to global health that increases the burden of liver diseases. Autophagy and apoptosis are important in Cd-induced liver injury. However, the regulatory mechanisms involved in the progression of Cd-induced liver damage are poorly understood. Herein, we investigated the role of vacuolar protein sorting 41 (VPS41) in Cd-induced autophagy and apoptosis in hepatocytes. We used targeted VPS41 regulation to elucidate the mechanism of Cd-induced hepatotoxicity. Our data showed that Cd triggered incomplete autophagy by downregulating VPS41, aggravating Cd-induced hepatocyte apoptosis. Mechanistically, Cd-induced VPS41 downregulation interfered with the mTORC1-TFEB/TFE3 axis, leading to an imbalance in autophagy initiation and termination and abnormal activation of autophagy. Moreover, Cd-induced downregulation of VPS41 inhibited autophagosome-lysosome fusion, leading to blocked autophagic flux. This triggers incomplete autophagy, which causes excessive P62 accumulation, accelerating Caspase-9 (CASP9) cleavage. Incomplete autophagy blocks clearance of cleaved CASP9 (CL-CASP9) via the autophagic pathway, promoting apoptosis. Notably, VPS41 overexpression alleviated Cd-induced incomplete autophagy and apoptosis, independent of the homotypic fusion and protein sorting complex. This study provides a new mechanistic understanding of the relationship between autophagy and apoptosis, suggesting that VPS41 is a new therapeutic target for Cd-induced liver damage.
Collapse
Affiliation(s)
- Tao Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Lianqi Yan
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha 410000, Hunan, China; Department of Orthopedics, Clinical Medical College of Yangzhou University, Subei People's Hospital, Yangzhou 225009, Jiangsu, China
| | - Li Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Jian Sun
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Huayi Qu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Yonggang Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Ruilong Song
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Xishuai Tong
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Jiangsu Key Laboratory of Zoonosis, Yangzhou 225009, Jiangsu, China
| | - Jiaqiao Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Yan Yuan
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Jianhong Gu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China.
| |
Collapse
|
20
|
Chen Q, Li L, Zhao J, Zhang Y, Xue X. Graphene oxide had adverse effects on sperm motility and morphology through oxidative stress. Toxicol In Vitro 2023; 92:105653. [PMID: 37487874 DOI: 10.1016/j.tiv.2023.105653] [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: 01/29/2023] [Revised: 07/15/2023] [Accepted: 07/21/2023] [Indexed: 07/26/2023]
Abstract
Graphene oxide (GO) is a new type of graphene material, but its effects on the male reproductive system are unclear. Here, we investigated the effects of GO on human sperm in vitro. Sperms were incubated with various doses of GO (0, 10, 20, or 40 μg/mL) for different times (1, 3, or 6 h) at 37 °C, followed by analyses of the sperm motility, viability, abnormalities, and DNA fragmentations. GO exposure significantly decreased sperm motility and viability, increased sperm abnormalities, and DNA fragmentation. Moreover, GO exposure resulted in a significant reduction of sperm mitochondrial membrane potential (MMP), which was confirmed by the ultrastructural changes of chromatin and mitochondria caused by GO. These data revealed the adverse effects of GO on sperm. Further research showed that GO exposure led to a significant increase in malondialdehyde (MDA) and reactive oxygen species (ROS) in sperm cells and a significant decrease in total superoxide dismutase (T-SOD) and glutathione peroxidase (GSH-Px). In addition, western blot analysis showed that the levels of Nrf-2 and HO-1 protein expression in GO-treated sperm cells were significantly increased compared to the control. These results indicated that GO had adverse effects on human sperm through oxidative stress, which was associated with Nrf-2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Qing Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Lei Li
- School of Materials Science and Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Jinyan Zhao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Yan Zhang
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China
| | - Xiang Xue
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China.
| |
Collapse
|
21
|
Chen JL, Wu X, Yin D, Jia XH, Chen X, Gu ZY, Zhu XM. Autophagy inhibitors for cancer therapy: Small molecules and nanomedicines. Pharmacol Ther 2023; 249:108485. [PMID: 37406740 DOI: 10.1016/j.pharmthera.2023.108485] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/27/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023]
Abstract
Autophagy is a conserved process in which the cytosolic materials are degraded and eventually recycled for cellular metabolism to maintain homeostasis. The dichotomous role of autophagy in pathogenesis is complicated. Accumulating reports have suggested that cytoprotective autophagy is responsible for tumor growth and progression. Autophagy inhibitors, such as chloroquine (CQ) and hydroxychloroquine (HCQ), are promising for treating malignancies or overcoming drug resistance in chemotherapy. With the rapid development of nanotechnology, nanomaterials also show autophagy-inhibitory effects or are reported as the carriers delivering autophagy inhibitors. In this review, we summarize the small-molecule compounds and nanomaterials inhibiting autophagic flux as well as the mechanisms involved. The nanocarrier-based drug delivery systems for autophagy inhibitors and their distinct advantages are also described. The progress of autophagy inhibitors for clinical applications is finally introduced, and their future perspectives are discussed.
Collapse
Affiliation(s)
- Jian-Li Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Xuan Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Dan Yin
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Xiao-Hui Jia
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Xu Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Ze-Yun Gu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Xiao-Ming Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, China.
| |
Collapse
|
22
|
Czarniewska E, Sielicki K, Maślana K, Mijowska E. In vivo study on borophene nanoflakes interaction with Tenebrio molitor beetle: viability of hemocytes and short-term immunity effect. Sci Rep 2023; 13:11823. [PMID: 37479709 PMCID: PMC10361989 DOI: 10.1038/s41598-023-38595-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 07/11/2023] [Indexed: 07/23/2023] Open
Abstract
The family of graphene-based materials welcomed a new member, borophene, in 2014. Research on synthesis routes and experimental study on physicochemical and biological (especially in vivo) properties still is strongly desired in order to evaluate its practical potential as a drug delivery-system. The effect of two-dimensional borophene nanoflakes on cells, systems and the entire animal organism has not been studied so far. Therefore, we investigated in vivo its biocompatibility with hemocytes in the Tenebrio molitor as a model organism. Short-term studies demonstrated that borophene nanoflakes at doses of 0.5, 1 or 2 µg of nanoflakes per insect did not induce hemocytotoxicity. Hemocytes exposed to nanoflakes showed morphology, adhesiveness and ability to form filopodia as in the control hemocytes. A detailed study indicates that borophene nanoflakes do not: (i) generate intracellular reactive oxygen species in hemocytes, (ii) affect the mitochondrial membrane potential and (iii) interfere with phagocytosis. Therefore, this contribution presents new in vivo insights into the group of two-dimensional materials which are one of the most promising materials for biomedical applications owing to their special structure and unique properties. However, long-term studies in insects and other animals are still necessary to confirm that borophene is biocompatible and biologically safe.
Collapse
Affiliation(s)
- Elżbieta Czarniewska
- Department of Animal Physiology and Developmental Biology, Institute of Experimental Biology, Faculty of Biology, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego Str. 6, 61-614, Poznan, Poland.
| | - Krzysztof Sielicki
- Faculty of Chemical Technology and Engineering, Nanomaterials Physicochemistry Department, West Pomeranian University of Technology, Szczecin, Piastow Ave. 42, 71-065, Szczecin, Poland
| | - Klaudia Maślana
- Faculty of Chemical Technology and Engineering, Nanomaterials Physicochemistry Department, West Pomeranian University of Technology, Szczecin, Piastow Ave. 42, 71-065, Szczecin, Poland
| | - Ewa Mijowska
- Faculty of Chemical Technology and Engineering, Nanomaterials Physicochemistry Department, West Pomeranian University of Technology, Szczecin, Piastow Ave. 42, 71-065, Szczecin, Poland.
| |
Collapse
|
23
|
Yuan J, Mo Y, Zhang Y, Zhang Y, Zhang Q. Nickel nanoparticles induce autophagy and apoptosis via HIF-1α/mTOR signaling in human bronchial epithelial cells. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 329:121670. [PMID: 37080518 PMCID: PMC10231338 DOI: 10.1016/j.envpol.2023.121670] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/31/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023]
Abstract
With the rapid development of nanotechnology, the potential adverse health effects of nanoparticles have been caught more attention and become global concerns. However, the underlying mechanisms in metal nanoparticle-induced toxic effects are still largely obscure. In this study, we investigated whether exposure to nickel nanoparticles (Nano-Ni) and titanium dioxide nanoparticles (Nano-TiO2) would alter autophagy and apoptosis levels in normal human bronchial epithelial BEAS-2B cells and the underlying mechanisms involved in this process. Our results showed that the expressions of autophagy- and apoptosis-associated proteins were dysregulated in cells exposed to Nano-Ni. However, exposure to the same doses of Nano-TiO2 had no significant effects on these proteins. In addition, exposure to Nano-Ni, but not Nano-TiO2, led to nuclear accumulation of HIF-1α and decreased phosphorylation of mTOR in BEAS-2B cells. Inhibition of HIF-1α by CAY10585 abolished Nano-Ni-induced decreased phosphorylation of mTOR, while activation of mTOR by MHY1485 did not affect Nano-Ni-induced nuclear accumulation of HIF-1α. Furthermore, both HIF-1α inhibition and mTOR activation abolished Nano-Ni-induced autophagy but enhanced Nano-Ni-induced apoptosis. Blockage of autophagic flux by Bafilomycin A1 exacerbated Nano-Ni-induced apoptosis, while activation of autophagy by Rapamycin effectively rescued Nano-Ni-induced apoptosis. In conclusion, our results demonstrated that Nano-Ni exposure caused increased levels of autophagy and apoptosis via the HIF-1α/mTOR signaling axis. Nano-Ni-induced autophagy has a protective role against Nano-Ni-induced apoptosis. These findings provide us with further insight into Nano-Ni-induced toxicity.
Collapse
Affiliation(s)
- Jiali Yuan
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, 40202, USA
| | - Yiqun Mo
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, 40202, USA
| | - Yue Zhang
- Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Yuanbao Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, 40202, USA
| | - Qunwei Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
24
|
Ristic B, Bosnjak M, Misirkic Marjanovic M, Stevanovic D, Janjetovic K, Harhaji-Trajkovic L. The Exploitation of Lysosomes in Cancer Therapy with Graphene-Based Nanomaterials. Pharmaceutics 2023; 15:1846. [PMID: 37514033 PMCID: PMC10383369 DOI: 10.3390/pharmaceutics15071846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/17/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Graphene-based nanomaterials (GNMs), including graphene, graphene oxide, reduced graphene oxide, and graphene quantum dots, may have direct anticancer activity or be used as nanocarriers for antitumor drugs. GNMs usually enter tumor cells by endocytosis and can accumulate in lysosomes. This accumulation prevents drugs bound to GNMs from reaching their targets, suppressing their anticancer effects. A number of chemical modifications are made to GNMs to facilitate the separation of anticancer drugs from GNMs at low lysosomal pH and to enable the lysosomal escape of drugs. Lysosomal escape may be associated with oxidative stress, permeabilization of the unstable membrane of cancer cell lysosomes, release of lysosomal enzymes into the cytoplasm, and cell death. GNMs can prevent or stimulate tumor cell death by inducing protective autophagy or suppressing autolysosomal degradation, respectively. Furthermore, because GNMs prevent bound fluorescent agents from emitting light, their separation in lysosomes may enable tumor cell identification and therapy monitoring. In this review, we explain how the characteristics of the lysosomal microenvironment and the unique features of tumor cell lysosomes can be exploited for GNM-based cancer therapy.
Collapse
Affiliation(s)
- Biljana Ristic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Dr. Subotića 1, 11000 Belgrade, Serbia
| | - Mihajlo Bosnjak
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Dr. Subotića 1, 11000 Belgrade, Serbia
| | - Maja Misirkic Marjanovic
- Department of Neurophysiology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Despot Stefan Blvd. 142, 11000 Belgrade, Serbia
| | - Danijela Stevanovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Dr. Subotića 1, 11000 Belgrade, Serbia
| | - Kristina Janjetovic
- Department of Neurophysiology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Despot Stefan Blvd. 142, 11000 Belgrade, Serbia
| | - Ljubica Harhaji-Trajkovic
- Department of Neurophysiology, Institute for Biological Research "Siniša Stanković"-National Institute of Republic of Serbia, University of Belgrade, Despot Stefan Blvd. 142, 11000 Belgrade, Serbia
| |
Collapse
|
25
|
Taheriazam A, Abad GGY, Hajimazdarany S, Imani MH, Ziaolhagh S, Zandieh MA, Bayanzadeh SD, Mirzaei S, Hamblin MR, Entezari M, Aref AR, Zarrabi A, Ertas YN, Ren J, Rajabi R, Paskeh MDA, Hashemi M, Hushmandi K. Graphene oxide nanoarchitectures in cancer biology: Nano-modulators of autophagy and apoptosis. J Control Release 2023; 354:503-522. [PMID: 36641122 DOI: 10.1016/j.jconrel.2023.01.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/08/2023] [Accepted: 01/09/2023] [Indexed: 01/16/2023]
Abstract
Nanotechnology is a growing field, with many potential biomedical applications of nanomedicine for the treatment of different diseases, particularly cancer, on the horizon. Graphene oxide (GO) nanoparticles can act as carbon-based nanocarriers with advantages such as a large surface area, good mechanical strength, and the capacity for surface modification. These nanostructures have been extensively used in cancer therapy for drug and gene delivery, photothermal therapy, overcoming chemotherapy resistance, and for imaging procedures. In the current review, we focus on the biological functions of GO nanoparticles as regulators of apoptosis and autophagy, the two major forms of programmed cell death. GO nanoparticles can either induce or inhibit autophagy in cancer cells, depending on the conditions. By stimulating autophagy, GO nanocarriers can promote the sensitivity of cancer cells to chemotherapy. However, by impairing autophagy flux, GO nanoparticles can reduce cell survival and enhance inflammation. Similarly, GO nanomaterials can increase ROS production and induce DNA damage, thereby sensitizing cancer cells to apoptosis. In vitro and in vivo experiments have investigated whether GO nanomaterials show any toxicity in major body organs, such as the brain, liver, spleen, and heart. Molecular pathways, such as ATG, MAPK, JNK, and Akt, can be regulated by GO nanomaterials, leading to effects on autophagy and apoptosis. These topics are discussed in this review to shed some lights towards the biomedical potential of GO nanoparticles and their biocompatibility, paving the way for their future application in clinical trials.
Collapse
Affiliation(s)
- Afshin Taheriazam
- Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Ghazaleh Gholamiyan Yousef Abad
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Shima Hajimazdarany
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Hassan Imani
- Department of Clinical Science, Faculty of Veterinary Medicine, Islamic Azad University, Shahr-e kord Branch, Chaharmahal and Bakhtiari, Iran
| | - Setayesh Ziaolhagh
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa; Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Vice President at Translational Sciences, Xsphera Biosciences Inc., 6 Tide Street, Boston, MA, 02210, USA
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkey
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Turkey
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Romina Rajabi
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | - Mahshid Deldar Abad Paskeh
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
26
|
Dai D, Chen Y, Wang Q, Wang C, Zhang C. Graphene oxide induced dynamic changes of autophagy-lysosome pathway and cell apoptosis via TFEB dysregulation in F98 cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 246:114172. [PMID: 36244172 DOI: 10.1016/j.ecoenv.2022.114172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/21/2022] [Accepted: 10/07/2022] [Indexed: 06/16/2023]
Abstract
The extensive application of graphene oxide (GO) nanomaterials increases the risk of their release into the environment, thus posing a threat to the human body. Multiple studies indicate that GO could lead to neurotoxicity, while the intricate biological effects of GO in astrocytes remain unclear. The autophagic disorder was considered an important part of the exposure risk of GO in the application of neuromedicine. This study explored the key regulators mediating the autophagic process in rat astroglioma-derived F98 cells caused by GO, especially the dynamic changes in the cellular physiological state over time. We identified transcription factor EB (TFEB), a critical regulator of the autophagy-lysosome pathway (ALP), as a crucial factor in GO-induced autophagy flux blockade and cell apoptosis. Specifically, the prolonged exposure to GO increased the amount of its cellular internalization, which gradually prevented TFEB from entering the nucleus, thereby leading to the subsequent ALP dysfunction and excessive cell apoptosis. Furthermore, STIP1 homology and U-Box containing protein 1 (STUB1), an E3 ubiquitin ligase, was responsible for GO-triggered TFEB dysregulation, and overexpression of STUB1 helped alleviate GO cytotoxicity. Our study highlights that impaired TFEB activity underlies compromised autophagy flux in GO-induced apoptosis and opens up new avenues for the application of GO-based nanotherapeutics with specific autophagy-regulating properties in the central nervous system.
Collapse
Affiliation(s)
- Danni Dai
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yuming Chen
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Qiuyu Wang
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Chunlin Wang
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Chao Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China.
| |
Collapse
|
27
|
Huang R, Chen J, Tan Q, Hu W, Chen X, Yu Y, Zang G, Tang Z. Role of Autophagy in the Ubiquitinated Hepatitis B Virus Core Antigen Enhancing Dendritic Cell Function. Viral Immunol 2022; 35:629-639. [DOI: 10.1089/vim.2022.0062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Run Huang
- Department of Infectious Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Chen
- Department of Infectious Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Quanhui Tan
- Department of Infectious Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiwei Hu
- Department of Infectious Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohua Chen
- Department of Infectious Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongsheng Yu
- Department of Infectious Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoqing Zang
- Department of Infectious Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenghao Tang
- Department of Infectious Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Dabrowski B, Zuchowska A, Brzozka Z. Graphene oxide internalization into mammalian cells – a review. Colloids Surf B Biointerfaces 2022; 221:112998. [DOI: 10.1016/j.colsurfb.2022.112998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/26/2022] [Accepted: 10/29/2022] [Indexed: 11/07/2022]
|
29
|
Mundekkad D, Cho WC. Mitophagy Induced by Metal Nanoparticles for Cancer Treatment. Pharmaceutics 2022; 14:2275. [PMID: 36365094 PMCID: PMC9699542 DOI: 10.3390/pharmaceutics14112275] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/12/2022] [Accepted: 10/19/2022] [Indexed: 11/30/2022] Open
Abstract
Research on nanoparticles, especially metal nanoparticles, in cancer therapy is gaining momentum. The versatility and biocompatibility of metal nanoparticles make them ideal for various applications in cancer therapy. They can bring about apoptotic cell death in cancer cells. In addition to apoptosis, nanoparticles mediate a special type of autophagy facilitated through mitochondria called mitophagy. Interestingly, nanoparticles with antioxidant properties are capable of inducing mitophagy by altering the levels of reactive oxygen species and by influencing signaling pathways like PINK/Parkin pathway and P13K/Akt/mTOR pathway. The current review presents various roles of metal nanoparticles in inducing mitophagy in cancer cells. We envision this review sheds some light on the blind spots in the research related to mitophagy induced by nanoparticles for cancer treatment.
Collapse
Affiliation(s)
- Deepa Mundekkad
- Centre for NanoBioTechnology (CNBT), Vellore Institute of Technology, Vellore 632014, India
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong SAR, China
| |
Collapse
|
30
|
Rahimi S, Chen Y, Zareian M, Pandit S, Mijakovic I. Cellular and subcellular interactions of graphene-based materials with cancerous and non-cancerous cells. Adv Drug Deliv Rev 2022; 189:114467. [PMID: 35914588 DOI: 10.1016/j.addr.2022.114467] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 01/24/2023]
Abstract
Despite significant advances in early detection and personalized treatment, cancer is still among the leading causes of death globally. One of the possible anticancer approaches that is presently receiving a lot of attention is the development of nanocarriers capable of specific and efficient delivery of anticancer drugs. Graphene-based materials are promising nanocarriers in this respect, due to their high drug loading capacity and biocompatibility. In this review, we present an overview on the interactions of graphene-based materials with normal mammalian cells at the molecular level as well as cellular and subcellular levels, including plasma membrane, cytoskeleton, and membrane-bound organelles such as lysosomes, mitochondria, nucleus, endoplasmic reticulum, and peroxisome. In parallel, we assemble the knowledge about the interactions of graphene-based materials with cancerous cells, that are considered as the potential applications of these materials for cancer therapy including metastasis treatment, targeted drug delivery, and differentiation to non-cancer stem cells. We highlight the influence of key parameters, such as the size and surface chemistry of graphene-based materials that govern the efficiency of internalization and biocompatibility of these particles in vitro and in vivo. Finally, this review aims to correlate the key parameters of graphene-based nanomaterials specially graphene oxide, such as size and surface modifications, to their interactions with the cancerous and non-cancerous cells for designing and engineering them for bio-applications and especially for therapeutic purposes.
Collapse
Affiliation(s)
- Shadi Rahimi
- Department of Biology and Biological Engineering, Chalmers University of Technology, Göteborg 41296, Sweden.
| | - Yanyan Chen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Göteborg 41296, Sweden
| | - Mohsen Zareian
- Department of Biology and Biological Engineering, Chalmers University of Technology, Göteborg 41296, Sweden; State Key Laboratory of Bio-based Material and Green Paper-making, Qilu University of Technology, Jinan, China
| | - Santosh Pandit
- Department of Biology and Biological Engineering, Chalmers University of Technology, Göteborg 41296, Sweden
| | - Ivan Mijakovic
- Department of Biology and Biological Engineering, Chalmers University of Technology, Göteborg 41296, Sweden; The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
31
|
Sousa D, Pereira SS, Pignatelli D. Modulation of Autophagy in Adrenal Tumors. Front Endocrinol (Lausanne) 2022; 13:937367. [PMID: 35966083 PMCID: PMC9373848 DOI: 10.3389/fendo.2022.937367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/15/2022] [Indexed: 01/18/2023] Open
Abstract
Adrenal masses are one of the most common tumors in humans. The majority are benign and non-functioning and therefore do not require immediate treatment. In contrast, the rare adrenal malignant tumors are often highly aggressive and with poor prognosis. Besides usually being detected in advanced stages, often already with metastases, one of the reasons of the unfavorable outcome of the patients with adrenal cancer is the absence of effective treatments. Autophagy is one of the intracellular pathways targeted by several classes of chemotherapeutics. Mitotane, the most commonly used drug for the treatment of adrenocortical carcinoma, was recently shown to also modulate autophagy. Autophagy is a continuous programmed cellular process which culminates with the degradation of cellular organelles and proteins. However, being a dynamic mechanism, understanding the autophagic flux can be highly complex. The role of autophagy in cancer has been described paradoxically: initially described as a tumor pro-survival mechanism, different studies have been showing that it may result in other outcomes, namely in tumor cell death. In adrenal tumors, this dual role of autophagy has also been addressed in recent years. Studies reported both induction and inhibition of autophagy as a treatment strategy of adrenal malignancies. Importantly, most of these studies were performed using cell lines. Consequently clinical studies are still required. In this review, we describe what is known about the role of autophagy modulation in treatment of adrenal tumors. We will also highlight the aspects that need further evaluation to understand the paradoxical role of autophagy in adrenal tumors.
Collapse
Affiliation(s)
- Diana Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Cancer Signaling & Metabolism Group, IPATIMUP- Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - Sofia S. Pereira
- Unidade Multidisciplinar de Investigação Biomédica (UMIB), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
- ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
| | - Duarte Pignatelli
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Cancer Signaling & Metabolism Group, IPATIMUP- Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- Department of Endocrinology, Centro Hospitalar e Universitário de S. João, Porto, Portugal
- Department of Biomedicine, Faculty of Medicine of the University of Porto, Porto, Portugal
| |
Collapse
|
32
|
Zhou X, Jin W, Sun H, Li C, Jia J. Perturbation of autophagy: An intrinsic toxicity mechanism of nanoparticles. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 823:153629. [PMID: 35131247 DOI: 10.1016/j.scitotenv.2022.153629] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/11/2022] [Accepted: 01/29/2022] [Indexed: 06/14/2023]
Abstract
Nanoparticles (NPs) have been widely used for various purposes due to their unique physicochemical properties. Such widespread applications greatly increase the possibility of human exposure to NPs in various ways. Once entering the human body, NPs may interfere with cellular homeostasis and thus affect the physiological system. As a result, it is necessary to evaluate the potential disturbance of NPs to multiple cell functions, including autophagy. Autophagy is an important cell function to maintain cellular homeostasis, and minimizing the disturbance caused by NP exposures to autophagy is critical to nanosafety. Herein, we summarized the recent research progress in nanotoxicity with particular focuses on the perturbation of NPs to cell autophagy. The basic processes of autophagy and complex relationships between autophagy and major human diseases were further discussed to emphasize the importance of keeping autophagy under control. Moreover, the most recent advances on perturbation of different types of NPs to autophagy were also reviewed. Last but not least, we also discussed major research challenges and potential coping strategies and proposed a safe-by-design strategy towards safer applications of NPs.
Collapse
Affiliation(s)
- Xiaofei Zhou
- College of Science & Technology, Hebei Agricultural University, Huanghua 061100, China
| | - Weitao Jin
- College of Science & Technology, Hebei Agricultural University, Huanghua 061100, China
| | - Hainan Sun
- Shandong Vocational College of Light Industry, Zibo 255300, China
| | - Chengjun Li
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China.
| | - Jianbo Jia
- Institute of Environmental Research at Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China.
| |
Collapse
|
33
|
Yang L, Hu L, Tang H, Chen X, Liu X, Zhang Y, Wen Y, Yang Y, Geng Y. The disruption of human trophoblast functions by autophagy activation through PI3K/AKT/mTOR pathway induced by exposure to titanium carbide (Ti 3C 2) MXene. Food Chem Toxicol 2022; 165:113128. [PMID: 35569596 DOI: 10.1016/j.fct.2022.113128] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 04/27/2022] [Accepted: 05/06/2022] [Indexed: 10/18/2022]
Abstract
Ti3C2 MXene, as a novel nanomaterial, has attracted great attention due to its promising properties in biomedical applications. However, the potential effects of Ti3C2 MXene on trophoblast functions have not been investigated. Here, we found that Ti3C2 MXene exposure weakened the extension ability of villus explants in vitro. We employed human trophoblast HTR-8/SVneo cells to reveal the underlying molecular mechanisms by which Ti3C2 MXene exposure affected trophoblast functions. Results showed that Ti3C2 MXene entered cells and mostly deposited in the cytoplasm, inhibiting cell migration and invasion abilities. Furthermore, we found that Ti3C2 MXene exposure elevated autophagy through the inhibition of the PI3K/AKT/mTOR pathway. Meanwhile, the application of an autophagy inhibitor (3-MA) prevented autophagy and restored cell viability, resulting in the recovery of cell migration and invasion abilities. These indicated that the cellular dysfunction induced by Ti3C2 MXene may be mediated by autophagy activation. Our results indicated that autophagy is a key factor in eliciting HTR-8/SVneo dysfunction after Ti3C2 MXene exposure, which could therefore damage placental development. Autophagy inhibition is a potential therapeutic strategy for alleviating the placental toxicity of nanoparticles.
Collapse
Affiliation(s)
- Limei Yang
- School of Public Health and Management, Chongqing Medical University, Chongqing, China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Le Hu
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Key Laboratory of Gynecologic Oncology of Gansu Province, Lanzhou, China
| | - Hongyu Tang
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China; College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Xuemei Chen
- School of Public Health and Management, Chongqing Medical University, Chongqing, China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Xueqing Liu
- School of Public Health and Management, Chongqing Medical University, Chongqing, China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Yue Zhang
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China; College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Yixian Wen
- School of Public Health and Management, Chongqing Medical University, Chongqing, China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Yongxiu Yang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Key Laboratory of Gynecologic Oncology of Gansu Province, Lanzhou, China.
| | - Yanqing Geng
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China; College of Basic Medicine, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
34
|
Lian CY, Chu BX, Xia WH, Wang ZY, Fan RF, Wang L. Persistent activation of Nrf2 in a p62-dependent non-canonical manner aggravates lead-induced kidney injury by promoting apoptosis and inhibiting autophagy. J Adv Res 2022; 46:87-100. [PMID: 37003700 PMCID: PMC10105071 DOI: 10.1016/j.jare.2022.04.016] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 04/10/2022] [Accepted: 04/25/2022] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Lead (Pb) is an environmental toxicant that poses severe health risks to humans and animals, especially renal disorders. Pb-induced nephrotoxicity has been attributed to oxidative stress, in which apoptosis and autophagy are core events. OBJECTIVES Nuclear factor erythroid 2-related factor 2 (Nrf2) acts as a major contributor to counteract oxidative damage, while hyperactivation or depletion of Nrf2 pathway can cause the redox imbalance to induce tissue injury. This study was performed to clarify the function and mechanism of Nrf2 in Pb-triggered kidney injury. METHODS AND RESULTS First, data showed that Pb exposure activates Nrf2 pathway in primary rat proximal tubular cells. Next, Pb-induced Nrf2 activation was effectively regulated by pharmacological modulation or siRNA-mediated knockdown in vitro and in vivo assays. Notably, Pb-triggered cytotoxicity, renal injury and concomitant apoptosis were improved by Nrf2 downregulation, confirming that Pb-induced persistent Nrf2 activation contributes to nephrotoxicity. Additionally, Pb-triggered autophagy blockage was relieved by Nrf2 downregulation. Mechanistically, we found that Pb-induced persistent Nrf2 activation is attributed to reduced Nrf2 ubiquitination and nuclear-cytoplasmic loss of Keap1 in a p62-dependent manner. CONCLUSIONS In conclusion, these findings highlight the dark side of persistent Nrf2 activation and potential crosstalk among Pb-induced persistent Nrf2 activation, apoptosis and autophagy blockage in Pb-triggered nephrotoxicity.
Collapse
Affiliation(s)
- Cai-Yu Lian
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China
| | - Bing-Xin Chu
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Wei-Hao Xia
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China
| | - Zhen-Yong Wang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China
| | - Rui-Feng Fan
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China.
| | - Lin Wang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China.
| |
Collapse
|
35
|
Tilija Pun N, Lee N, Song SH, Jeong CH. Pitavastatin Induces Cancer Cell Apoptosis by Blocking Autophagy Flux. Front Pharmacol 2022; 13:854506. [PMID: 35387352 PMCID: PMC8977529 DOI: 10.3389/fphar.2022.854506] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/28/2022] [Indexed: 11/25/2022] Open
Abstract
Statins, a class of lipid-lowering drugs, are used in drug repositioning for treatment of human cancer. However, the molecular mechanisms underlying statin-induced cancer cell death and autophagy are not clearly defined. In the present study, we showed that pitavastatin could increase apoptosis in a FOXO3a-dependent manner in the oral cancer cell line, SCC15, and the colon cancer cell line, SW480, along with the blockade of autophagy flux. The inhibition of autophagy by silencing the LC3B gene reduced apoptosis, while blockade of autophagy flux using its inhibitor, Bafilomycin A1, further induced apoptosis upon pitavastatin treatment, which suggested that autophagy flux blockage was the cause of apoptosis by pitavastatin. Further, the FOXO3a protein accumulated due to the blockade of autophagy flux which in turn was associated with the induction of ER stress by transcriptional upregulation of PERK-CHOP pathway, subsequently causing apoptosis due to pitavastatin treatment. Taken together, pitavastatin-mediated blockade of autophagy flux caused an accumulation of FOXO3a protein, thereby leading to the induction of PERK, ultimately causing CHOP-mediated apoptosis in cancer cells. Thus, the present study highlighted the additional molecular mechanism underlying the role of autophagy flux blockade in inducing ER stress, eventually leading to apoptosis by pitavastatin.
Collapse
Affiliation(s)
- Nirmala Tilija Pun
- College of Pharmacy, Keimyung University, Daegu, South Korea.,Boston Children's Hospital, Boston, MA, United States
| | - Naeun Lee
- College of Pharmacy, Keimyung University, Daegu, South Korea
| | - Sang-Hoon Song
- College of Pharmacy, Keimyung University, Daegu, South Korea
| | - Chul-Ho Jeong
- College of Pharmacy, Keimyung University, Daegu, South Korea
| |
Collapse
|
36
|
Gong JY, Holt MG, Hoet PHM, Ghosh M. Neurotoxicity of four frequently used nanoparticles: a systematic review to reveal the missing data. Arch Toxicol 2022; 96:1141-1212. [DOI: 10.1007/s00204-022-03233-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/20/2022] [Indexed: 12/27/2022]
|
37
|
Zhang Q, Cao S, Qiu F, Kang N. Incomplete autophagy: Trouble is a friend. Med Res Rev 2022; 42:1545-1587. [PMID: 35275411 DOI: 10.1002/med.21884] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 01/26/2022] [Accepted: 02/22/2022] [Indexed: 01/18/2023]
Abstract
Incomplete autophagy is an impaired self-eating process of intracellular macromolecules and organelles in which accumulated autophagosomes do not fuse with lysosomes for degradation, resulting in the blockage of autophagic flux. In this review, we summarized the literature over the past decade describing incomplete autophagy, and found that different from the double-edged sword effect of general autophagy on promoting cell survival or death, incomplete autophagy plays a crucial role in disrupting cellular homeostasis, and promotes only cell death. What matters is that incomplete autophagy is closely relevant to the pathogenesis and progression of various human diseases, which, meanwhile, intimately linking to the pharmacologic and toxicologic effects of several compounds. Here, we comprehensively reviewed the latest progress of incomplete autophagy on molecular mechanisms and signaling pathways. Moreover, implications of incomplete autophagy for pharmacotherapy are also discussed, which has great relevance for our understanding of the distinctive role of incomplete autophagy in cellular physiology and disease. Consequently, targeting incomplete autophagy may contribute to the development of novel generation therapeutic agents for diverse human diseases.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Biochemistry, School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Shijie Cao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Feng Qiu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.,Department of Medicinal Chemistry, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Ning Kang
- Department of Biochemistry, School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| |
Collapse
|
38
|
Targeting autophagy, oxidative stress, and ER stress for neurodegenerative diseases treatment. J Control Release 2022; 345:147-175. [DOI: 10.1016/j.jconrel.2022.03.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/27/2022] [Accepted: 03/01/2022] [Indexed: 12/13/2022]
|
39
|
Paskeh MDA, Entezari M, Clark C, Zabolian A, Ranjbar E, Farahani MV, Saleki H, Sharifzadeh SO, Far FB, Ashrafizadeh M, Samarghandian S, Khan H, Ghavami S, Zarrabi A, Łos MJ. Targeted regulation of autophagy using nanoparticles: New insight into cancer therapy. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166326. [DOI: 10.1016/j.bbadis.2021.166326] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/31/2021] [Accepted: 12/11/2021] [Indexed: 12/12/2022]
|
40
|
Pan X, Cheng D, Ruan C, Hong Y, Lin C. Development of Graphene-Based Materials in Bone Tissue Engineaering. GLOBAL CHALLENGES (HOBOKEN, NJ) 2022; 6:2100107. [PMID: 35140982 PMCID: PMC8812920 DOI: 10.1002/gch2.202100107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/31/2021] [Indexed: 06/14/2023]
Abstract
Bone regeneration-related graphene-based materials (bGBMs) are increasingly attracting attention in tissue engineering due to their special physical and chemical properties. The purpose of this review is to quantitatively analyze mass academic literature in the field of bGBMs through scientometrics software CiteSpace, to demonstrate the rules and trends of bGBMs, thus to analyze and summarize the mechanisms behind the rules, and to provide clues for future research. First, the research status, hotspots, and frontiers of bGBMs are analyzed in an intuitively and vividly visualized way. Next, the extracted important subjects such as fabrication techniques, cytotoxicity, biodegradability, and osteoinductivity of bGBMs are presented, and the different mechanisms, in turn, are also discussed. Finally, photothermal therapy, which is considered an emerging area of application of bGBMs, is also presented. Based on this approach, this work finds that different studies report differing opinions on the biological properties of bGBMS due to the lack of consistency of GBMs preparation. Therefore, it is necessary to establish more standards in fabrication, characterization, and testing for bGBMs to further promote scientific progress and clinical translation.
Collapse
Affiliation(s)
- Xiaoling Pan
- College of StomatologyXinjiang Medical UniversityUrumqiXinjiang830011P. R. China
- Department of Oral Maxillofacial SurgeryShenzhen HospitalSouthern Medical UniversityShenzhen518000P. R. China
| | - Delin Cheng
- Research Center for Human Tissue and Organs DegenerationInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
| | - Changshun Ruan
- Research Center for Human Tissue and Organs DegenerationInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
| | - Yonglong Hong
- Department of Oral Maxillofacial SurgeryShenzhen HospitalSouthern Medical UniversityShenzhen518000P. R. China
| | - Cheng Lin
- Department of Oral Maxillofacial SurgeryShenzhen HospitalSouthern Medical UniversityShenzhen518000P. R. China
| |
Collapse
|
41
|
A Four-Step Cascade Drug-Release Management Strategy for Transcatheter Arterial Chemoembolization (TACE) Therapeutic Applications. Polymers (Basel) 2021; 13:polym13213701. [PMID: 34771257 PMCID: PMC8588239 DOI: 10.3390/polym13213701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 10/24/2021] [Indexed: 11/21/2022] Open
Abstract
The purpose of this study was to develop a four-step cascade drug-release system for transcatheter arterial chemoembolization (TACE) therapeutic applications according to disease-driven and patient-focused design theories. The four steps underlying these strategies involve the blockage of nutrient supply, nanoparticles, codelivery and the cell cytotoxic effect. Calibrated spherical gellan gum (GG) and nanoparticle-containing gellan gum microspheres were prepared using a water-in-oil emulsification method. Self-assembled nanoparticles featuring amine-functionalized graphene oxide (AFGO) as the doxorubicin (Dox) carrier were prepared. The results confirm that, as a drug carrier, AFGO–Dox nanoparticles can facilitate the transport of doxorubicin into HepG2 liver cancer cells. Subsequently, AFGO–Dox was introduced into gellan gum (GG) microspheres, thus forming GG/AFGO–Dox microspheres with a mean size of 200–700 μm. After a drug release experiment lasting 28 days, the amount of doxorubicin released from 674 and 226 μm GG/AFGO–Dox microspheres was 2.31 and 1.18 μg/mg, respectively. GG/AFGO–Dox microspheres were applied in a rabbit ear embolization model, where ischemic necrosis was visible on the ear after 12 days. Our aim for the future is to provide better embolization agents for transcatheter arterial chemoembolization (TACE) using this device.
Collapse
|
42
|
Ristic B, Harhaji-Trajkovic L, Bosnjak M, Dakic I, Mijatovic S, Trajkovic V. Modulation of Cancer Cell Autophagic Responses by Graphene-Based Nanomaterials: Molecular Mechanisms and Therapeutic Implications. Cancers (Basel) 2021; 13:cancers13164145. [PMID: 34439299 PMCID: PMC8392723 DOI: 10.3390/cancers13164145] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/06/2021] [Accepted: 08/13/2021] [Indexed: 01/18/2023] Open
Abstract
Simple Summary Graphene-based nanomaterials (GNM) are one-to-several carbon atom-thick flakes of graphite with at least one lateral dimension <100 nm. The unique electronic structure, high surface-to-volume ratio, and relatively low toxicity make GNM potentially useful in cancer treatment. GNM such as graphene, graphene oxide, graphene quantum dots, and graphene nanofibers are able to induce autophagy in cancer cells. During autophagy the cell digests its own components in organelles called lysosomes, which can either kill cancer cells or promote their survival, as well as influence the immune response against the tumor. However, a deeper understanding of GNM-autophagy interaction at the mechanistic and functional level is needed before these findings could be exploited to increase GNM effectiveness as cancer therapeutics and drug delivery systems. In this review, we analyze molecular mechanisms of GNM-mediated autophagy modulation and its possible implications for the use of GNM in cancer therapy. Abstract Graphene-based nanomaterials (GNM) are plausible candidates for cancer therapeutics and drug delivery systems. Pure graphene and graphene oxide nanoparticles, as well as graphene quantum dots and graphene nanofibers, were all able to trigger autophagy in cancer cells through both transcriptional and post-transcriptional mechanisms involving oxidative/endoplasmic reticulum stress, AMP-activated protein kinase, mechanistic target of rapamycin, mitogen-activated protein kinase, and Toll-like receptor signaling. This was often coupled with lysosomal dysfunction and subsequent blockade of autophagic flux, which additionally increased the accumulation of autophagy mediators that participated in apoptotic, necrotic, or necroptotic death of cancer cells and influenced the immune response against the tumor. In this review, we analyze molecular mechanisms and structure–activity relationships of GNM-mediated autophagy modulation, its consequences for cancer cell survival/death and anti-tumor immune response, and the possible implications for the use of GNM in cancer therapy.
Collapse
Affiliation(s)
- Biljana Ristic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (B.R.); (I.D.)
| | - Ljubica Harhaji-Trajkovic
- Department of Neurophysiology, Institute for Biological Research “Sinisa Stankovic”, National Institute of Republic of Serbia, University of Belgrade, 11060 Belgrade, Serbia;
| | - Mihajlo Bosnjak
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Ivana Dakic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (B.R.); (I.D.)
| | - Srdjan Mijatovic
- Clinic for Emergency Surgery, Clinical Centre of Serbia, 11000 Belgrade, Serbia;
| | - Vladimir Trajkovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (B.R.); (I.D.)
- Correspondence:
| |
Collapse
|
43
|
Xiaoli F, Yaqing Z, Ruhui L, Xuan L, Aijie C, Yanli Z, Chen H, Lili C, Longquan S. Graphene oxide disrupted mitochondrial homeostasis through inducing intracellular redox deviation and autophagy-lysosomal network dysfunction in SH-SY5Y cells. JOURNAL OF HAZARDOUS MATERIALS 2021; 416:126158. [PMID: 34492938 DOI: 10.1016/j.jhazmat.2021.126158] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 05/05/2021] [Accepted: 05/16/2021] [Indexed: 06/13/2023]
Abstract
Graphene oxide (GO) nanomaterials have significant advantages for drug delivery and electrode materials in neural science, however, their exposure risks to the central nervous system (CNS) and toxicity concerns are also increased. The current studies of GO-induced neurotoxicity remain still ambiguous, let alone the mechanism of how complicated GO chemistry affects its biological behavior with neural cells. In this study, we characterized the commercially available GO in detail and investigated its biological adverse effects using cultured SH-SY5Y cells. We found that ultrasonic processing in medium changed the oxidation status and surface reactivity on the planar surface of GO due to its hydration activity, causing lipid peroxidation and cell membrane damage. Subsequently, ROS-disrupted mitochondrial homeostasis, resulting from the activation of NOX2 signaling, was observed following GO internalization. The autophagy-lysosomal network was initiated as a defensive reaction to obliterate oxidative damaged mitochondria and foreign nanomaterials, which was ineffective due to reduced lysosomal degradation capacity. These sequential cellular responses exacerbated mitochondrial stress, leading to apoptotic cell death. These data highlight the importance of the structure-related activity of GO on its biological properties and provide an in-depth understanding of how GO-derived cellular redox signaling induces mitochondrion-related cascades that modulate cell functionality and survival.
Collapse
Affiliation(s)
- Feng Xiaoli
- Stomatology Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou 510515, China
| | - Zhang Yaqing
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Luo Ruhui
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lai Xuan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Chen Aijie
- Stomatology Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhang Yanli
- Stomatology Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hu Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Chen Lili
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shao Longquan
- Stomatology Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou 510515, China.
| |
Collapse
|
44
|
Liu G, Lai D, Jiang Y, Yang H, Zhao H, Zhang Y, Liu D, Pang Y. Demethylzeylasteral Exerts Antitumor Effects via Disruptive Autophagic Flux and Apoptotic Cell Death in Human Colorectal Cancer Cells and Increases Cell Chemosensitivity to 5-Fluorouracil. Anticancer Agents Med Chem 2021; 22:851-863. [PMID: 34102996 DOI: 10.2174/1871520621666210608104021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/23/2021] [Accepted: 04/12/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Demethylzeylasteral (ZST93), a pharmacologically active triterpenoid monomer extracted from Tripterygium wilfordii Hook F (TWHF), has been reported to exert antineoplastic effects in several cancer cell types. However, the anti-tumour effects of ZST93 in human colorectal cancer (CRC) cells are unknown. OBJECTIVE The aim of the present study was to evaluate the antitumor effects of ZST93 on cell cycle arrest, disruptive autophagic flux, apoptotic cell death, and enhanced chemosensitivity to 5-FU in humans CRC cells. METHODS The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide(MTT) assay, colony formation assay, flow cytometry, immunoblotting, immunofluorescence, 5-ethynyl-20-deoxyuridine (EdU) incorporation assay, and autophagy analysis were used to evaluate the effects of ZST93 on cell viability, cell cycle progression, apoptosis and autophagy in two human CRC cell lines. Moreover, ZST93's combined anti-tumour effects with 5-fluorouracil (5-FU) were evaluated. RESULTS ZST93 inhibited CRC cell proliferation and growth. It was responsible for blocked cell cycle transition by arresting CRC cells in the G0/G1 phase via down-regulation of CDK4, CDK6, Cyclin D1, and c-MYC. Moreover, ZST93 induced suppressive autophagic flux and caspase-3-dependent cell death, which were further strengthened by the blocking of the autophagy process using chloroquine (CQ). Moreover, ZST93 enhanced CRC cells' chemosensitivity to 5-FU via modulation of autophagy and apoptosis. CONCLUSION ZST93 exerts anti-tumour effects via disruptive autophagic flux and apoptotic cell death in human CRC cells and increases cell chemosensitivity to 5-FU. These results provide insights into the utilisation of ZST93 as an adjuvant or direct autophagy inhibitor and suggest ZST93 as a novel therapeutic strategy for treating CRC.
Collapse
Affiliation(s)
- Guiyuan Liu
- The Affiliated Hospital of Chongqing Three Gorges Medical College, Chongqing, China
| | - Dengxiang Lai
- The Affiliated Hospital of Chongqing Three Gorges Medical College, Chongqing, China
| | - Yi Jiang
- The Affiliated Hospital of Chongqing Three Gorges Medical College, Chongqing, China
| | - Hongjing Yang
- Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing Three Gorges Medical College, Chongqing, China
| | - Hui Zhao
- The Affiliated Hospital of Chongqing Three Gorges Medical College, Chongqing, China
| | - Yonghui Zhang
- Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing Three Gorges Medical College, Chongqing, China
| | - Dan Liu
- Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing Three Gorges Medical College, Chongqing, China
| | - Yi Pang
- Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing Three Gorges Medical College, Chongqing, China
| |
Collapse
|
45
|
Yu Y, Men S, Zhang Y. miR-20a-5p ameliorates ovalbumin (OVA)-induced mouse model of allergic asthma through targeting ATG7-regulated cell death, fibrosis and inflammation. Int Immunopharmacol 2021; 95:107342. [PMID: 33684878 DOI: 10.1016/j.intimp.2020.107342] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 12/09/2020] [Accepted: 12/23/2020] [Indexed: 01/12/2023]
Abstract
Autophagy plays an essential role in modulating asthma progression. MiR-20a-5p can regulate autophagy, but its effects on allergic asthma are still unclear. The aim of this study was to explore the potential of miR-20a-5p on autophagy-modulated airway remodeling and to reveal the underlying molecular mechanisms. We found that miR-20a-5p expression was markedly down-regulated in lung of ovalbumin (OVA)-induced mouse model with allergic asthma and in cells stimulated by OVA. Meanwhile, autophagy, apoptosis, fibrosis and inflammatory response were detected in pulmonary tissues from OVA-treated mice. Importantly, luciferase assays showed that ATG7 was a target of miR-20a-5p. We also found that miR-20a-5p over-expression markedly reduced ATG7, while its inhibition promoted ATG7 in cells. In addition, over-expressing miR-20a-5p in OVA-treated cells significantly decreased ATG7 expression levels, along with markedly reduced autophagy, apoptotic cell death, fibrosis and inflammatory response. These results were similar to the effects of autophagy inhibitor 3-Methyladenine (3-MA), indicating that miR-20a-5p was involved in autophagy-induced apoptosis, fibrosis and inflammation. In vivo experiments further demonstrated that miR-20a-5p over-expression was associated with ATG7 reduction in parallel with the alleviated airway remodeling in OVA-treated mice also through suppressing collagen accumulation, apoptosis and inflammation. Similarly, animal studies further confirmed that miR-20a-5p functioned as an autophagy inhibitor to mitigate allergic asthma development. Therefore, miR-20a-5p may be a promising biomarker and therapeutic target during asthma progression by regulating ATG7-modulated autophagy.
Collapse
Affiliation(s)
- Yanyan Yu
- Department of Pediatric Asthma, Lianyungang Maternal and Child Health Hospital in Jiangsu Province, Jiangsu 215003, China.
| | - Shuai Men
- Department of Respiratory Specialty the Children's Hospital of Suzhou University, Jiangsu 215003, China
| | - Yuhong Zhang
- Department of Respiratory Specialty the Children's Hospital of Suzhou University, Jiangsu 215003, China
| |
Collapse
|
46
|
Guo W, Chen Z, Feng X, Shen G, Huang H, Liang Y, Zhao B, Li G, Hu Y. Graphene oxide (GO)-based nanosheets with combined chemo/photothermal/photodynamic therapy to overcome gastric cancer (GC) paclitaxel resistance by reducing mitochondria-derived adenosine-triphosphate (ATP). J Nanobiotechnology 2021; 19:146. [PMID: 34011375 PMCID: PMC8136184 DOI: 10.1186/s12951-021-00874-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/27/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Paclitaxel (PTX) has been suggested to be a promising front-line drug for gastric cancer (GC), while P-glycoprotein (P-gp) could lead to drug resistance by pumping PTX out of GC cells. Consequently, it might be a hopeful way to combat drug resistance by inhibiting the out-pumping function of P-gp. RESULTS In this study, we developed a drug delivery system incorporating PTX onto polyethylene glycol (PEG)-modified and oxidized sodium alginate (OSA)-functionalized graphene oxide (GO) nanosheets (NSs), called PTX@GO-PEG-OSA. Owing to pH/thermal-sensitive drug release properties, PTX@GO-PEG-OSA could induced more obvious antitumor effects on GC, compared to free PTX. With near infrared (NIR)-irradiation, PTX@GO-PEG-OSA could generate excessive reactive oxygen species (ROS), attack mitochondrial respiratory chain complex enzyme, reduce adenosine-triphosphate (ATP) supplement for P-gp, and effectively inhibit P-gp's efflux pump function. Since that, PTX@GO-PEG-OSA achieved better therapeutic effect on PTX-resistant GC without evident toxicity. CONCLUSIONS In conclusion, PTX@GO-PEG-OSA could serve as a desirable strategy to reverse PTX's resistance, combined with chemo/photothermal/photodynamic therapy.
Collapse
Affiliation(s)
- Weihong Guo
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Zhian Chen
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Xiaoli Feng
- Guangdong Provincial Stomatology Hospital, Southern Medical University, Guangzhou, 510000 China
| | - Guodong Shen
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Huilin Huang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Yanrui Liang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Bingxia Zhao
- Guangdong Provincial Key Laboratory of Cancer Immunotherapy, Guangzhou Key Laboratory of Tumor Immunology Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515 PR China
| | - Guoxin Li
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Yanfeng Hu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| |
Collapse
|
47
|
Xavier Mendes A, Moraes Silva S, O'Connell CD, Duchi S, Quigley AF, Kapsa RMI, Moulton SE. Enhanced Electroactivity, Mechanical Properties, and Printability through the Addition of Graphene Oxide to Photo-Cross-linkable Gelatin Methacryloyl Hydrogel. ACS Biomater Sci Eng 2021; 7:2279-2295. [PMID: 33956434 DOI: 10.1021/acsbiomaterials.0c01734] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The human tissues most sensitive to electrical activity such as neural and muscle tissues are relatively soft, and yet traditional conductive materials used to interface with them are typically stiffer by many orders of magnitude. Overcoming this mismatch, by creating both very soft and electroactive materials, is a major challenge in bioelectronics and biomaterials science. One strategy is to imbue soft materials, such as hydrogels, with electroactive properties by adding small amounts of highly conductive nanomaterials. However, electroactive hydrogels reported to date have required relatively large volume fractions (>1%) of added nanomaterial, have shown only modest electroactivity, and have not been processable via additive manufacturing to create 3D architectures. Here, we describe the development and characterization of improved biocompatible photo-cross-linkable soft hybrid electroactive hydrogels based on gelatin methacryloyol (GelMA) and large area graphene oxide (GO) flakes, which resolve each of these three limitations. The addition of very small amounts (less than a 0.07% volume fraction) of GO to a 5% w/v GelMA hydrogel resulted in a dramatic (∼35-fold) decrease in the impedance at 1 Hz compared with GelMA alone. The GelMA/GO coated indium tin oxide (ITO) electrode also showed a considerable reduction in the impedance at 1 kHz (down to 170 Ω compared with 340 Ω for the GelMA-coated ITO), while charge injection capacity increased more than 6-fold. We attribute this enhanced electroactivity to the increased electroactive surface area contributed by the GO. Despite this dramatic change in electroactivity, the GelMA/GO composite hydrogels' mechanical properties were only moderately affected. Mechanical properties increased by ∼2-fold, and therefore, the hydrogels' desired softness of <4 kPa was retained. Also, we demonstrate how light attenuation through the gel can be used to create a stiffness gradient with the exposed surface of the gel having an elastic modulus of <1.5 kPa. GO addition also enhanced the rheological properties of the GelMA composites, thus facilitating 3D extrusion printing. GelMA/GO enhanced filament formation as well as improved printability and the shape fidelity/integrity of 3D printed structures compared with GelMA alone. Additionally, the GelMA/GO 3D printed structures presented a higher electroactive behavior than nonprinted samples containing the same GelMA/GO amount, which can be attributed to the higher electroactive surface area of 3D printed structures. These findings provide new rational choices of electroactive hydrogel (EAH) compositions with broad potential applications in bioelectronics, tissue engineering, and drug delivery.
Collapse
Affiliation(s)
- Alexandre Xavier Mendes
- ARC Centre of Excellence for Electromaterials Science, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, Victoria 3122, Australia.,The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia
| | - Saimon Moraes Silva
- ARC Centre of Excellence for Electromaterials Science, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, Victoria 3122, Australia.,The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia
| | - Cathal D O'Connell
- School of Electrical and Biomedical Engineering, RMIT University, Melbourne, Victoria 3001, Australia.,The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia
| | - Serena Duchi
- The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia.,Department of Medicine, University of Melbourne, St Vincent's Hospital, Melbourne, Victoria 3065, Australia
| | - Anita F Quigley
- School of Electrical and Biomedical Engineering, RMIT University, Melbourne, Victoria 3001, Australia.,The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia.,Department of Medicine, University of Melbourne, St Vincent's Hospital, Melbourne, Victoria 3065, Australia
| | - Robert M I Kapsa
- ARC Centre of Excellence for Electromaterials Science, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, Victoria 3122, Australia.,School of Electrical and Biomedical Engineering, RMIT University, Melbourne, Victoria 3001, Australia.,The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia.,Department of Medicine, University of Melbourne, St Vincent's Hospital, Melbourne, Victoria 3065, Australia
| | - Simon E Moulton
- ARC Centre of Excellence for Electromaterials Science, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, Victoria 3122, Australia.,The Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Melbourne, Victoria 3065, Australia.,Iverson Health Innovation Research Institute, Swinburne University of Technology, Melbourne, Victoria 3122, Australia
| |
Collapse
|
48
|
Liu S, Xu A, Gao Y, Xie Y, Liu Z, Sun M, Mao H, Wang X. Graphene oxide exacerbates dextran sodium sulfate-induced colitis via ROS/AMPK/p53 signaling to mediate apoptosis. J Nanobiotechnology 2021; 19:85. [PMID: 33766052 PMCID: PMC7995754 DOI: 10.1186/s12951-021-00832-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 03/14/2021] [Indexed: 12/30/2022] Open
Abstract
Background Graphene oxide (GO), a novel carbon-based nanomaterial, has promising applications in biomedicine. However, it induces potential cytotoxic effects on the gastrointestinal (GI) tract cells, and these effects have been largely uncharacterized. The present study aimed to explore the toxic effects of GO on the intestinal tract especially under pre-existing inflammatory conditions, such as inflammatory bowel disease (IBD), and elucidate underlying mechanisms. Results Our findings indicated that oral gavage of GO worsened acute colitis induced by 2.5% dextran sodium sulfate (DSS) in mice. In vitro, GO exacerbated DSS-induced inflammation and apoptosis in the FHC cell line, an ideal model of intestinal epithelial cells (IECs). Further, the potential mechanism underlying GO aggravated mice colitis and cell inflammation was explored. Our results revealed that GO treatment triggered apoptosis in FHC cells through the activation of reactive oxygen species (ROS)/AMP-activated protein kinase (AMPK)/p53 pathway, as evidenced by the upregulation of cytochrome c (Cytc), Bax, and cleaved caspase-3 (c-cas3) and the downregulation of Bcl-2. Interestingly, pretreatment with an antioxidant, N-acetyl-L-cysteine, and a specific inhibitor of AMPK activation, Compound C (Com.C), effectively inhibited GO-induced apoptosis in FHC cells. Conclusions Our data demonstrate that GO-induced IECs apoptosis via ROS/AMPK/p53 pathway activation accounts for the exacerbation of colitis in vivo and aggravation of inflammation in vitro. These findings provide a new insight into the pathogenesis of IBD induced by environmental factors. Furthermore, our findings enhance our understanding of GO as a potential environmental toxin, which helps delineate the risk of exposure to patients with disturbed intestinal epithelial barrier/inflammatory disorders such as IBD. ![]()
Collapse
Affiliation(s)
- Siliang Liu
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Angao Xu
- Huizhou Medicine Institute, Huizhou, 516003, People's Republic of China
| | - Yanfei Gao
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Yue Xie
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Zhipeng Liu
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Meiling Sun
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Hua Mao
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Xinying Wang
- Department of Gastroenterology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
49
|
Dai D, He L, Chen Y, Zhang C. Astrocyte responses to nanomaterials: Functional changes, pathological changes and potential applications. Acta Biomater 2021; 122:66-81. [PMID: 33326883 DOI: 10.1016/j.actbio.2020.12.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/30/2020] [Accepted: 12/08/2020] [Indexed: 12/15/2022]
Abstract
Astrocytes are responsible for regulating and optimizing the functional environment of neurons in the brain and can reduce the adverse impacts of external factors by protecting neurons. However, excessive astrocyte activation upon stimulation may alter their initial protective effect and actually lead to aggravation of injury. Similar to the dual effects of astrocytes in the response to injury within the central nervous system (CNS), nanomaterials (NMs) can have either toxic or beneficial effects on astrocytes, serving to promote injury or inhibit tumors. As the important physiological functions of astrocytes have been gradually revealed, the effects of NMs on astrocytes and the underlying mechanisms have become a new frontier in nanomedicine and neuroscience. This review summarizes the in vitro and in vivo findings regarding the effects of various NMs on astrocytes, focusing on functional alterations and pathological processes in astrocytes, as well as the possible underlying mechanisms. We also emphasize the importance of co-culture models in studying the interaction between NMs and cells of the CNS. Finally, we discuss NMs that have shown promise for application in astrocyte-related diseases and propose some challenges and suggestions for further investigations, with the aim of providing guidance for the widespread application of NMs in the CNS.
Collapse
Affiliation(s)
- Danni Dai
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Longwen He
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yuming Chen
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Chao Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China.
| |
Collapse
|
50
|
Zhang Y, Zhang Y, Wu J, Liu J, Kang Y, Hu C, Feng X, Liu W, Luo H, Chen A, Chen L, Shao L. Effects of carbon-based nanomaterials on vascular endothelia under physiological and pathological conditions: interactions, mechanisms and potential therapeutic applications. J Control Release 2021; 330:945-962. [DOI: 10.1016/j.jconrel.2020.10.067] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 10/31/2020] [Accepted: 10/31/2020] [Indexed: 12/11/2022]
|