1
|
Milenkovska R, Geskovski N, Shalabalija D, Mihailova L, Makreski P, Lukarski D, Stojkovski I, Simonoska Crcarevska M, Mladenovska K. Radiosensitizing properties of dual-functionalized carbon nanostructures loaded with temozolomide. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2025; 16:229-251. [PMID: 39995757 PMCID: PMC11849551 DOI: 10.3762/bjnano.16.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 01/16/2025] [Indexed: 02/26/2025]
Abstract
In the present study, temozolomide (TMZ), a drug used for the treatment of anaplastic astrocytoma and glioblastoma multiforme (GBM), was incorporated into multiwalled carbon nanotubes (MWCNTs) and a MWCNTs-graphene (MWCNTs-G) hybrid compound, covalently functionalized with polyethylene glycol (PEG) 6000 and folic acid (FA), with an aim to prepare nanocarriers with the potential to prolong the drug circulation time, cross the blood-brain-tumor barrier (BBTB), and provide targeted and controlled drug release in the brain tumor cells. Cytotoxicity and effects on cell membrane integrity of the blank and TMZ-loaded dual-functionalized carbon nanostructures (CNs) were evaluated in vitro on a GBM cell line (U87MG), as well as their radiosensitizing properties after exposure of the pre-treated GBM cells to gamma radiation with a standard clinical dose for patients with GBM. All prepared formulations underwent biopharmaceutical and physicochemical characterization, including the formulations exposed to irradiation under the same conditions. For physicochemical characterization of the formulations, different techniques were used by which successful functionalization of the CNs and TMZ loading were confirmed and visualized; no significant changes in the structure of the CNs and TMZ after irradiation were observed. With single and dual functionalization, formulations with relatively high TMZ loading efficiency and drug content were prepared. They exhibited homogeneous particle size distributions and mean particle sizes and surface charges suitable for crossing the BBTB and targeting brain cancer cells. A biphasic drug release profile was observed for all functionalized TMZ-loaded formulations in simulated in vivo conditions, with a sustained release pointing to the potential for controlled release of TMZ in brain tumor cells. The formulations of the hybrid CN MWCNTs-G compared to the corresponding MWCNTs were characterized by a similar or slightly higher TMZ content, larger particle size, similar surface charge, and slightly faster TMZ release, which can be attributed to the planar structure of graphene that promotes TMZ binding to the surface on a larger scale. For the irradiated CNs, lower values for particle size, more positive values for surface charge, and accelerated TMZ release were observed, which could be explained by changes in the physicochemical characteristics of the prepared formulations upon irradiation. Significant concentration-dependent toxicity was observed for blank dual-functionalized CNs, being higher for MWCNTs-G-PEG6000-FA compared to MWCNTs-PEG6000-FA at the same formulation concentrations. With incorporation of TMZ into the functionalized CNs, the cell viability additionally decreased, maintaining the trend for higher cytotoxicity of the hybrid CN. Additional decrease in the viability of cells was observed when GBM cells pre-treated with the corresponding CNs were exposed to irradiation, which could be ascribed to changes in size, surface charge, and release kinetics of TMZ and to irradiation-induced changes in the microenvironment and cell membranes that promote uptake of a larger volume of carriers in the GBM cells. The higher cytotoxicity observed in the hybrid carrier formulations could most likely be attributed to the length of the hybrid carrier and the higher proportion of planar surface, which promotes more intense contact with the cells and rupture of cell membranes. Overall, the findings demonstrate the radiosensitizing properties of not only TMZ but also of CNs and point to a clinical benefit from combined treatment with carbon nanocarriers of TMZ and radiotherapy in GBM.
Collapse
Affiliation(s)
- Radmila Milenkovska
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, Ss. Cyril and Methodius University in Skopje, Blv. Mother Theresa No. 45, 1000 Skopje, Republic of North Macedonia
| | - Nikola Geskovski
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, Ss. Cyril and Methodius University in Skopje, Blv. Mother Theresa No. 45, 1000 Skopje, Republic of North Macedonia
| | - Dushko Shalabalija
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, Ss. Cyril and Methodius University in Skopje, Blv. Mother Theresa No. 45, 1000 Skopje, Republic of North Macedonia
| | - Ljubica Mihailova
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, Ss. Cyril and Methodius University in Skopje, Blv. Mother Theresa No. 45, 1000 Skopje, Republic of North Macedonia
| | - Petre Makreski
- Institute of Chemistry, Faculty of Natural Sciences and Mathematics, Ss. Cyril and Methodius University in Skopje, Str. Arhimedova No. 5, 1000 Skopje, Republic of North Macedonia
| | - Dushko Lukarski
- University Clinic of Radiotherapy and Oncology, Faculty of Medicine, Ss. Cyril and Methodius University in Skopje, Blv. Mother Theresa No. 17, 1000 Skopje, Republic of North Macedonia
- Faculty of Medicine, Ss. Cyril and Methodius University in Skopje, Str. 50th Division No. 6, 1000 Skopje, Republic of North Macedonia
| | - Igor Stojkovski
- University Clinic of Radiotherapy and Oncology, Faculty of Medicine, Ss. Cyril and Methodius University in Skopje, Blv. Mother Theresa No. 17, 1000 Skopje, Republic of North Macedonia
- Faculty of Medicine, Ss. Cyril and Methodius University in Skopje, Str. 50th Division No. 6, 1000 Skopje, Republic of North Macedonia
| | - Maja Simonoska Crcarevska
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, Ss. Cyril and Methodius University in Skopje, Blv. Mother Theresa No. 45, 1000 Skopje, Republic of North Macedonia
| | - Kristina Mladenovska
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, Ss. Cyril and Methodius University in Skopje, Blv. Mother Theresa No. 45, 1000 Skopje, Republic of North Macedonia
| |
Collapse
|
2
|
Sharma M, Alessandro P, Cheriyamundath S, Lopus M. Therapeutic and diagnostic applications of carbon nanotubes in cancer: recent advances and challenges. J Drug Target 2024; 32:287-299. [PMID: 38252035 DOI: 10.1080/1061186x.2024.2309575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 01/11/2024] [Indexed: 01/23/2024]
Abstract
Carbon nanotubes (CNTs) are allotropes of carbon, composed of carbon atoms forming a tube-like structure. Their high surface area, chemical stability, and rich electronic polyaromatic structure facilitate their drug-carrying capacity. Therefore, CNTs have been intensively explored for several biomedical applications, including as a potential treatment option for cancer. By incorporating smart fabrication strategies, CNTs can be designed to specifically target cancer cells. This targeted drug delivery approach not only maximizes the therapeutic utility of CNTs but also minimizes any potential side effects of free drug molecules. CNTs can also be utilised for photothermal therapy (PTT) which uses photosensitizers to generate reactive oxygen species (ROS) to kill cancer cells, and in immunotherapeutic applications. Regarding the latter, for example, CNT-based formulations can preferentially target intra-tumoural regulatory T-cells. CNTs also act as efficient antigen presenters. With their capabilities for photoacoustic, fluorescent and Raman imaging, CNTs are excellent diagnostic tools as well. Further, metallic nanoparticles, such as gold or silver nanoparticles, are combined with CNTs to create nanobiosensors to measure biological reactions. This review focuses on current knowledge about the theranostic potential of CNT, challenges associated with their large-scale production, their possible side effects and important parameters to consider when exploring their clinical usage.
Collapse
Affiliation(s)
- Muskan Sharma
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidyanagari, Mumbai, India
| | - Parodi Alessandro
- Department of Translational Medicine, Sirius University of Science and Technology, Sirius, Russia
| | - Sanith Cheriyamundath
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidyanagari, Mumbai, India
| | - Manu Lopus
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidyanagari, Mumbai, India
| |
Collapse
|
3
|
Evariste L, Verneuil L, Silvestre J, Mouchet F, Gauthier L, Boutonnet JC, Flahaut E, Pinelli E. Cellular uptake of multi-walled carbon nanotubes is associated to genotoxic and teratogenic effects towards the freshwater diatom Nitzschia linearis. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 275:107067. [PMID: 39222567 DOI: 10.1016/j.aquatox.2024.107067] [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: 05/17/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
The increase in industrial production of multi-walled carbon nanotubes (MWCNTs) raises concerns about their potential adverse effects associated to environmental releases, especially in aquatic environments where they are likely to accumulate. This study focuses on the environmental impact of MWCNTs, specifically on a benthic freshwater diatom (Nitzschia linearis), which plays a major role in the primary production of water bodies. The obtained results indicate that exposure to MWCNTs in the presence of natural organic matter (NOM) inhibits diatom's growth in a dose-dependent manner after 72 h of exposure. Interestingly, the photosystem II quantum yield (PSIIQY) in diatoms remains unaffected even after exposure to MWCNTs at 10 mg/L. After 48 h of exposure, MWCNTs are found to bind preferentially to extracellular polymeric substances (EPS) produced by diatoms, which could decrease their toxicity by limiting their interaction with this organism. However, measurement of genotoxicity and teratogenicity in diatoms exposed to MWCNTs revealed that the exposure to MWCNTs increased the occurrence of cells with micronuclei and abnormal frustules. Microscopy analyses including two-photon excitation microscopy (TPEM) revealed the internalization of MWCNTs. Investigations of the diatom's frustule structure using Scanning electron microscopy (SEM) indicated that the presence of pore structures constitutes a pathway allowing MWCNTs uptake. The presence in the diatom's cytoplasm of MWCNTs might possibly induce disturbances of the cellular components, leading to the observed genotoxic and teratogenic effects. In view of previous studies, this work underscores the need for further studies on the interaction between nanomaterials and different diatom species, given the species-specific nature of the interactions.
Collapse
Affiliation(s)
- Lauris Evariste
- Centre de Recherche sur la Biodiversité et l'Environnement, UMR CNRS 5300, Castanet-Tolosan, France.
| | - Laurent Verneuil
- Centre de Recherche sur la Biodiversité et l'Environnement, UMR CNRS 5300, Castanet-Tolosan, France
| | - Jérôme Silvestre
- Centre de Recherche sur la Biodiversité et l'Environnement, UMR CNRS 5300, Castanet-Tolosan, France
| | - Florence Mouchet
- Centre de Recherche sur la Biodiversité et l'Environnement, UMR CNRS 5300, Castanet-Tolosan, France
| | - Laury Gauthier
- Centre de Recherche sur la Biodiversité et l'Environnement, UMR CNRS 5300, Castanet-Tolosan, France
| | | | - Emmanuel Flahaut
- CIRIMAT, Université Toulouse 3 Paul Sabatier, Toulouse INP, CNRS, Université de Toulouse, 118 Route de Narbonne cedex 9, 31062, Toulouse, France
| | - Eric Pinelli
- Centre de Recherche sur la Biodiversité et l'Environnement, UMR CNRS 5300, Castanet-Tolosan, France
| |
Collapse
|
4
|
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
|
5
|
García-Hevia L, Soltani R, González J, Chaloin O, Ménard-Moyon C, Bianco A, L. Fanarraga M. Carbon nanotubes targeted to the tumor microenvironment inhibit metastasis in a preclinical model of melanoma. Bioact Mater 2024; 34:237-247. [PMID: 38223536 PMCID: PMC10787223 DOI: 10.1016/j.bioactmat.2023.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 01/16/2024] Open
Abstract
Despite notable progress in cancer therapy, metastatic diseases continue to be the primary cause of cancer-related mortality. Multi-walled carbon nanotubes (MWCNTs) can enter tissues and cells and interfere with the dynamics of the cytoskeletal nanofilaments biomimetically. This endows them with intrinsic anti-tumoral effects comparable to those of microtubule-binding chemotherapies such as Taxol®. In this study, our focus was on exploring the potential of oxidized MWCNTs in selectively targeting the vascular endothelial growth factor receptor (VEGFR). Our objective was to evaluate their effectiveness in inhibiting metastatic growth by inducing anti-proliferative, anti-migratory, and cytotoxic effects on both cancer and tumor microenvironment cells. Our findings demonstrated a significant reduction of over 80 % in malignant melanoma lung metastases and a substantial enhancement in overall animal welfare following intravenous administration of the targeted biodegradable MWCNTs. Furthermore, the combination of these nanomaterials with the conventional chemotherapy agent Taxol® yielded a remarkable 90 % increase in the antimetastatic effect. These results highlight the promising potential of this combined therapeutic approach against metastatic disease and are of paramount importance as metastasis is responsible for nearly 60,000 deaths each year.
Collapse
Affiliation(s)
- Lorena García-Hevia
- The Nanomedicine Group, Universidad de Cantabria-IDIVAL, Avda Herrera Oria s/n, 39011, Santander, Spain
| | - Rym Soltani
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572, University of Strasbourg, ISIS, 67000, Strasbourg, France
| | - Jesús González
- The Nanomedicine Group, Universidad de Cantabria-IDIVAL, Avda Herrera Oria s/n, 39011, Santander, Spain
| | - Olivier Chaloin
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572, University of Strasbourg, ISIS, 67000, Strasbourg, France
| | - Cécilia Ménard-Moyon
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572, University of Strasbourg, ISIS, 67000, Strasbourg, France
| | - Alberto Bianco
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572, University of Strasbourg, ISIS, 67000, Strasbourg, France
| | - Mónica L. Fanarraga
- The Nanomedicine Group, Universidad de Cantabria-IDIVAL, Avda Herrera Oria s/n, 39011, Santander, Spain
| |
Collapse
|
6
|
Priyam J, Saxena U. Therapeutic applications of carbon nanomaterials in renal cancer. Biotechnol Lett 2023; 45:1395-1416. [PMID: 37864745 DOI: 10.1007/s10529-023-03429-0] [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: 06/07/2023] [Revised: 08/27/2023] [Accepted: 08/31/2023] [Indexed: 10/23/2023]
Abstract
Carbon nanomaterials (CNMs), including carbon nanotubes (CNTs), graphene, and nanodiamonds (NDs), have shown great promise in detecting and treating numerous cancers, including kidney cancer. CNMs can increase the sensitivity of diagnostic techniques for better kidney cancer identification and surveillance. They enable targeted medicine delivery specifically to tumour locations, with little effect on healthy tissue. Because of their unique chemical and physical characteristics, they can avoid the body's defence mechanisms, making it easier to accumulate where tumours exist. Consequently, CNMs provide more effective drug delivery to kidney cancer cells. It also helps in improving the efficacy of treatment. This review explores the potential of several CNMs in improving therapeutic strategies for kidney cancer. We briefly covered the physicochemical properties and therapeutic applications of CNMs. Additionally, we discussed how structural modifications in CNMs enhance their precision in treating renal cancer. A thorough overview of CNM-based gene, peptide, and drug delivery strategies for the treatment of renal cancer is presented in this review. It covers information on other CNM-based therapeutic approaches, such as hyperthermia, photodynamic therapy, and photoacoustic therapy. Also, the interactions of CNMs with the tumour microenvironment (TME) are explored, including modulation of the immune response, regulation of tumour hypoxia, interactions between CNMs and TME cells, effects of TME pH on CNMs, and more. Finally, potential side effects of CNMs, such as toxicity, bio corona formation, enzymatic degradation, and biocompatibility, are also discussed.
Collapse
Affiliation(s)
- Jyotsna Priyam
- Department of Biotechnology, National Institute of Technology Warangal, Warangal, Telangana, 506004, India
| | - Urmila Saxena
- Department of Biotechnology, National Institute of Technology Warangal, Warangal, Telangana, 506004, India.
| |
Collapse
|
7
|
Jiang Y, Zhu C, Huang H, Huang G, Fu B, Xi X. TUBA1C is a potential new prognostic biomarker and promotes bladder urothelial carcinoma progression by regulating the cell cycle. BMC Cancer 2023; 23:716. [PMID: 37528357 PMCID: PMC10391756 DOI: 10.1186/s12885-023-11209-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 07/23/2023] [Indexed: 08/03/2023] Open
Abstract
BACKGROUND TUBA1C is an α-tubulin isoform involved in mitosis, and its dysregulation has been implicated in tumor progression. There is still no clear understanding of its role in bladder urothelial carcinoma (BLCA). METHODS This study examined the differential expression of TUBA1C and its prognostic significance in bladder cancer based on data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) and also assessed the correlation of TUBA1C expression level with immune cell infiltration and immune checkpoint gene expression levels and the half-inhibitory concentration (IC50) of different chemotherapeutic agents. Immunotherapy response was estimated using the Tumor Immune Dysfunction and Exclusion (TIDE) algorithm. We detected TUBA1C expression in BLCA cells using PCR and Western blotting. Functional assays, including CCK-8, colony formation, transwell, apoptosis and cell cycle assays, were also performed to assess the oncogenic role of TUBA1C in BLCA. RESULT In three independent public cohorts, TUBA1C was significantly upregulated in bladder tumor tissues, and high TUBA1C expression in bladder cancer was associated with a poorer outcome than low expression. TUBA1C was an independent prognostic risk factor for bladder cancer, and numerous immune checkpoint genes and infiltrating immune cells were associated with TUBA1C. TIDE analysis revealed that TUBA1C showed great potential for predicting the immunotherapy response in bladder cancer patients. In addition, drug sensitivity analysis revealed that high TUBA1C expression indicated sensitivity to multiple chemotherapeutic agents. Functional assays revealed that silencing TUBA1C significantly inhibited the proliferation, migration and invasion of BLCA cells and induced apoptosis and cell cycle arrest. CONCLUSION The overexpression of TUBA1C in bladder cancer predicts a poor prognosis and may also be a potential immunotherapeutic target. As a prognostic marker, TUBA1C influences tumor progression by regulating the cell cycle.
Collapse
Affiliation(s)
- Yi Jiang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chao Zhu
- Department of Blood Transfusion, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Haoxuan Huang
- Department of Urology, The Third Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gaomin Huang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bin Fu
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China.
| | - Xiaoqing Xi
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
8
|
Zhang H, Luo QQ, Hu ML, Wang N, Qi HZ, Zhang HR, Ding L. Discovery of potent microtubule-destabilizing agents targeting for colchicine site by virtual screening, biological evaluation, and molecular dynamics simulation. Eur J Pharm Sci 2023; 180:106340. [PMID: 36435355 DOI: 10.1016/j.ejps.2022.106340] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 11/03/2022] [Accepted: 11/22/2022] [Indexed: 11/24/2022]
Abstract
Microtubule has been considered as attractive therapeutic target for various cancers. Although numerous of chemically diverse compounds targeting to colchicine site have been reported, none of them was approved by Food and Drug Administration. In this investigation, the virtual screening methods, including pharmacophore model, molecular docking, and interaction molecular fingerprints similarity, were applied to discover novel microtubule-destabilizing agents from database with 324,474 compounds. 22 compounds with novel scaffolds were identified as microtubule-destabilizing agents, and then submitted to the biological evaluation. Among these 22 hits, hit4 with novel scaffold represents the best anti-proliferative activity with IC50 ranging from 4.51 to 14.81 μM on four cancer cell lines. The in vitro assays reveal that hit4 can effectively inhibit tubulin assembly, and disrupt the microtubule network in MCF-7 cell at a concentration-dependent manner. Finally, the molecular dynamics simulation analysis exhibits that hit4 can stably bind to colchicine site, interact with key residues, and induce αT5 and βT7 regions changes. The values of ΔGbind for the tubulin-colchicine and tubulin-hit4 are -172.9±10.5 and -166.0±12.6 kJ·mol-1, respectively. The above results indicate that the hit4 is a novel microtubule destabilizing agent targeting to colchicine-binding site, which could be developed as a promising tubulin polymerization inhibitor with higher activity for cancer therapy.
Collapse
Affiliation(s)
- Hui Zhang
- College of Life Science, Northwest Normal University, Lanzhou, Gansu 730070, PR China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Qing-Qing Luo
- College of Life Science, Northwest Normal University, Lanzhou, Gansu 730070, PR China
| | - Mei-Ling Hu
- College of Life Science, Northwest Normal University, Lanzhou, Gansu 730070, PR China
| | - Ni Wang
- College of Life Science, Northwest Normal University, Lanzhou, Gansu 730070, PR China
| | - Hua-Zhao Qi
- College of Life Science, Northwest Normal University, Lanzhou, Gansu 730070, PR China
| | - Hong-Rui Zhang
- College of Life Science, Northwest Normal University, Lanzhou, Gansu 730070, PR China
| | - Lan Ding
- College of Life Science, Northwest Normal University, Lanzhou, Gansu 730070, PR China
| |
Collapse
|
9
|
Pu Z, Wei Y, Sun Y, Wang Y, Zhu S. Carbon Nanotubes as Carriers in Drug Delivery for Non-Small Cell Lung Cancer, Mechanistic Analysis of Their Carcinogenic Potential, Safety Profiling and Identification of Biomarkers. Int J Nanomedicine 2022; 17:6157-6180. [PMID: 36523423 PMCID: PMC9744892 DOI: 10.2147/ijn.s384592] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/23/2022] [Indexed: 04/04/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is a global burden leading to millions of deaths worldwide every year. Nanomedicine refers to the use of materials at the nanoscale for drug delivery and subsequent therapeutic approaches in cancer. Carbon nanotubes (CNTs) are widely used as nanocarriers for therapeutic molecules such as plasmids, siRNAs, antisense agents, aptamers and molecules related to the immunotherapy for several cancers. They are usually functionalized and loaded with standard drug molecules to improve their therapeutic efficiency. Functionalization and drug loading possibly decrease the genotoxic and carcinogenic potential of CNTs. In addition, the targeted cytotoxic properties of the drug improve and undesired toxicity decreases after drug loading and/or conjugation with proteins, including antibodies. For intended drug delivery, a lysosomal pH of 5.5 is more suitable and effective for the slow and extended release of cytotoxic drugs than a physiological of pH 7.4. Remarkably, CNTs possess intrinsic antitumor properties and are usually internalized by endocytosis. After being internalized, several mechanisms are involved in the therapeutic and carcinogenic effects of CNTs. They are generally safe for therapy, and their toxicity profile remains dependent on their physicochemical properties. Moreover, the dose, route, duration of exposure, surface properties and degradative potential determine the toxicity outcomes of CNTs locally or systemically. In summary, the use of CNTs in drug delivery and NSCLC therapy, as well as their genotoxic and carcinogenic potential and the possible mechanisms, has been discussed in this review. The therapeutic index is generally high for NSCLC cells treated with drug-loaded CNTs; therefore, they are effective carriers in implementing targeted therapy for NSCLC.
Collapse
Affiliation(s)
- Zhongjian Pu
- Department of Oncology, Haian Hospital of Traditional Chinese Medicine, Haian, 216600, People’s Republic of China
| | - Yujia Wei
- School of Medicine, Yangzhou University, Yangzhou, 225009, People’s Republic of China
- Department of General Practice, Suzhou Wuzhong Hospital of Traditional Chinese Medicine, Suzhou, 215101, People’s Republic of China
| | - Yuanpeng Sun
- Department of Oncology, Haian Hospital of Traditional Chinese Medicine, Haian, 216600, People’s Republic of China
| | - Yajun Wang
- Department of Oncology, Haian Hospital of Traditional Chinese Medicine, Haian, 216600, People’s Republic of China
| | - Shilin Zhu
- Department of Oncology, Haian Hospital of Traditional Chinese Medicine, Haian, 216600, People’s Republic of China
| |
Collapse
|
10
|
Bora D, Sharma A, John SE, Shankaraiah N. Development of hydrazide hydrazone-tethered combretastatin-oxindole derivatives as antimitotic agents. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
11
|
RDIVpSGP motif of ASPP2 binds to 14-3-3 and enhances ASPP2/k18/14-3-3 ternary complex formulation to promote BRAF/MEK/ERK signal inhibited cell proliferation in hepatocellular carcinoma. Cancer Gene Ther 2022; 29:1616-1627. [PMID: 35504951 DOI: 10.1038/s41417-022-00474-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/27/2022] [Accepted: 04/08/2022] [Indexed: 02/04/2023]
Abstract
The Apoptosis Stimulating Protein of p53 2 (ASPP2) is a heterozygous insufficient tumor suppressor; however, its molecular mechanism(s) in tumor suppression is not completely understood. ASPP2 plays an essential role in cell growth, as shown by liver hepatocellular carcinoma (LIHC) RNA-seq assay using the Cancer Genome Atlas (TCGA) and High-Throughput-PCR assay using ASPP2 knockdown cells. These observations were further confirmed by in vivo and in vitro experiments. Mechanistically, N-terminus ASPP2 interacted with Keratin 18 (k18) in vivo and in vitro. Interestingly, the RDIVpSGP motif of ASPP2 associates with 14-3-3 and promotes ASPP2/k18/14-3-3 ternary-complex formation which promotes MEK/ERK signal activation by impairing 14-3-3 and BRAF association. Additionally, ASPP2-rAd injection promotes paclitaxel-suppressed tumor growth by suppressing cell proliferation in the BALB/c nude mice model. ASPP2 and k18 were preferentially downregulated in Hepatocellular Carcinoma (HCC), which predicted poor prognosis in HCC patients. Overall, these findings suggested that ASPP2 promoted BRAF/MEK/ERK signal activation by promoting the formation of an ASPP2/k18/14-3-3 ternary complex via the RDIVpSGP motif at the N terminus. Moreover, this study provides novel insights into the molecular mechanism of tumor suppression in HCC patients.
Collapse
|
12
|
Yadav K, Ali SA, Mohanty AK, Muthusamy E, Subaharan K, Kaul G. MSN, MWCNT and ZnO nanoparticle-induced CHO-K1 cell polarisation is linked to cytoskeleton ablation. J Nanobiotechnology 2021; 19:45. [PMID: 33579304 PMCID: PMC7881565 DOI: 10.1186/s12951-021-00779-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/19/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND The cellular response to nanoparticles (NPs) for the mechanical clue and biochemical changes are unexplored. Here, we provide the comprehensive analysis of the Chinese Hamster Ovary (CHO-K1) cell line to study cell behaviour following the exposure of mesoporous silica nanoparticle (MSN), multiwall carbon nanotubes (MWCNTs), and zinc oxide (ZnO) NPs. RESULTS Through the high-throughput proteomic study, we observed that the effect of NPs is alone not restricted to cell viability but also on cell polarisation. In the case of MSN, no drastic changes were observed in cellular morphology, but it upregulated chaperons that might prevent protein aggregation. However, MWCNT showed elongated cell appearance with numerous cytoplasmic vacuoles, and induce lamellipodia formation through actin polymerisation. The cytoskeleton remodelling was accompanied by the increased expression of Dlc-1, cofilin and Rac1 proteins. While ZnO NPs resulted in the rounded cell morphology along with nuclear abnormalities. The proteome analysis revealed that UBXN11 control cell roundness and DOCK3 leads to actin stress fibre formation and finally, loss of cell adhesion. It enhances the expression of catastrophic DNA damage and apoptotic proteins, which was unrecoverable even after 72 h, as confirmed by the colony formation assay. All three NPs trigger over-expression of the endocytic pathway, ubiquitination, and proteasomal complex proteins. The data indicate that ZnO and MSN entered into the cells through clathrin-mediated pathways; whereas, MWCNT invades through ER-mediated phagocytosis. CONCLUSIONS Based on the incubation and concentration of NPs, our work provides evidence for the activation of Rac-Rho signalling pathway to alter cytoskeleton dynamics. Our results assist as a sensitive early molecular readout for nanosafety assessment.
Collapse
Affiliation(s)
- Karmveer Yadav
- N.T. Lab-1, Division of Animal Biochemistry, ICAR-National Dairy Research Institute, Karnal, 132001, India.
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, 132001, Haryana, India
| | - Ashok Kumar Mohanty
- Cell Biology and Proteomics Lab, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, 132001, Haryana, India
| | - Eshwarmoorthy Muthusamy
- Chemistry and Physics of Materials Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - Kesavan Subaharan
- Division of Germplasm, Conservation and Utilisation, National Bureau of Agricultural Insect Resources, Bangalore, 560024, India
| | - Gautam Kaul
- N.T. Lab-1, Division of Animal Biochemistry, ICAR-National Dairy Research Institute, Karnal, 132001, India.
| |
Collapse
|