1
|
Dey S, Ghosh M, Dev A. Signalling and molecular pathways, overexpressed receptors of colorectal cancer and effective therapeutic targeting using biogenic silver nanoparticles. Gene 2025; 936:149099. [PMID: 39557372 DOI: 10.1016/j.gene.2024.149099] [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: 08/15/2024] [Revised: 10/18/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
Increasing morbidity and mortality in CRC is a potential threat to human health. The major challenges for better treatment outcomes are the heterogeneity of CRC cases, complicated molecular pathway cross-talks, the influence of gut dysbiosis in CRC, and the lack of multimodal target-specific drug delivery. The overexpression of many receptors in CRC cells may pave the path for targeting them with multiple ligands. The design of a more target-specific drug-delivery device with multiple ligand-functionalized, green-synthesized silver nanoparticles is highly promising and may also deliver other approved chemotherapeutic agents. This review presents the various aspects of colorectal cancer and over-expressed receptors that can be targeted with appropriate ligands to enhance the specific drug delivery potency of green synthesised silver nanoparticles. This review aims to broaden further research into this multi-ligand functionalised, safer and effective silver nano drug delivery system.
Collapse
Affiliation(s)
- Sandip Dey
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India
| | - Manik Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India
| | - Abhimanyu Dev
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India.
| |
Collapse
|
2
|
Pietsch FL, Haag F, Ayx I, Grawe F, Vellala AK, Schoenberg SO, Froelich MF, Tharmaseelan H. Textural heterogeneity of liver lesions in CT imaging - comparison of colorectal and pancreatic metastases. Abdom Radiol (NY) 2024; 49:4295-4306. [PMID: 39115682 PMCID: PMC11522118 DOI: 10.1007/s00261-024-04511-5] [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: 05/20/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 10/30/2024]
Abstract
PURPOSE Tumoral heterogeneity poses a challenge for personalized cancer treatments. Especially in metastasized cancer, it remains a major limitation for successful targeted therapy, often leading to drug resistance due to tumoral escape mechanisms. This work explores a non-invasive radiomics-based approach to capture textural heterogeneity in liver lesions and compare it between colorectal cancer (CRC) and pancreatic cancer (PDAC). MATERIALS AND METHODS In this retrospective single-center study 73 subjects (42 CRC, 31 PDAC) with 1291 liver metastases (430 CRC, 861 PDAC) were segmented fully automated on contrast-enhanced CT images by a UNet for medical images. Radiomics features were extracted using the Python package Pyradiomics. The mean coefficient of variation (CV) was calculated patient-wise for each feature to quantify the heterogeneity. An unpaired t-test identified features with significant differences in feature variability between CRC and PDAC metastases. RESULTS In both colorectal and pancreatic liver metastases, interlesional heterogeneity in imaging can be observed using quantitative imaging features. 75 second-order features were extracted to compare the varying textural characteristics. In total, 18 radiomics features showed a significant difference (p < 0.05) in their expression between the two malignancies. Out of these, 16 features showed higher levels of variability within the cohort of pancreatic metastases, which, as illustrated in a radar plot, suggests greater textural heterogeneity for this entity. CONCLUSIONS Radiomics has the potential to identify the interlesional heterogeneity of CT texture among individual liver metastases. In this proof-of-concept study for the quantification and comparison of imaging-related heterogeneity in liver metastases a variation in the extent of heterogeneity levels in CRC and PDAC liver metastases was shown.
Collapse
Affiliation(s)
- Friedrich L Pietsch
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Florian Haag
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Isabelle Ayx
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Freba Grawe
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Abhinay K Vellala
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Stefan O Schoenberg
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Matthias F Froelich
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Hishan Tharmaseelan
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| |
Collapse
|
3
|
Deng S, Liu Y, Liu X, Yu J, Chen Y, Huo J. Inhibition of colorectal cancer aggressiveness by Oleanolic acid through Nur77 degradation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156192. [PMID: 39520953 DOI: 10.1016/j.phymed.2024.156192] [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: 07/15/2024] [Revised: 10/17/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) is the second primary malignancy in China with tough treatment challenge. Although Oleanolic acid (OA) protects against various cancers, its mechanisms in CRC are not well defined. Our previously study showed that Nur77 has CRC promoting effect. Thus, we investigated the roles of OA as Nur77 ligand and the regulatory effects on Nur77 degradation in CRC progression. METHODS The proliferative and metastatic phenotypes of OA was examined by CCK-8, EdU, organoid culture, would healing and transwell assays, respectively. Epithelial-mesenchymal transition (EMT) properties were assessed by Western blotting (WB). The interaction between OA and Nur77 was monitored by molecular docking and Molecular Dynamics stimulation (MD). Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene set enrichment analysis (GSEA) were employed to screen the downstream regulatory pathways. The half-time and proteasome degradation of Nur77 were treated with cycloheximide (CHX) and MG132. Co-immunoprecipitation (Co-IP) and ubiquitination assays were employed to detect direct association between Nur77 and PPARγ. Rescued experiments were performed by Nur77 agonist Cytosporone B (Csn-B) treatment. The findings were verified in xenograft and in situ models. RESULTS For the first time, we found the effect of OA on ubiquitination degradation. OA inhibited CRC cell survival and EMT phenotypes by suppressing Nur77. Mechanistically, OA directly bind to Nur77 and facilitated the ubiquitin degradation of Nur77. During this process, PPARγ acted as the ubiquitination activator via interacting with Nur77. Rescued experiments revealed that OA-induced inhibition was recovered by replenishing Nur77. In both subcutaneous and orthotopic CRC models, OA exhibited significant anti-tumor effect together with Nur77 inhibition. CONCLUSION We revealed a new regulatory effect of OA in CRC tumorigenesis via PPARγ-mediated Nur77 ubiquitin degradation.
Collapse
Affiliation(s)
- Shan Deng
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.
| | - Yuping Liu
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Xiyu Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, People's Republic of China
| | - Jialin Yu
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Yan Chen
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China; Research Center for Multicomponent of Traditional Chinese Medicine and Microecology, Jiangsu Provincial Academy of Chinese Medicine, 100 Shizi Road, Nanjing, Jiangsu, 210028, People's Republic of China.
| | - Jiege Huo
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China; Department of Oncology, Jiangsu Province Academy of Traditional Chinese Medicine, 100 Shizi Road, Nanjing, Jiangsu, 210028, People's Republic of China.
| |
Collapse
|
4
|
Tian Y, Zhao Q, Wu H, Guo J, Wu H. VWA2 protein molecular mechanism predicts colorectal cancer: Promoting cell invasion and migration by inhibiting NK cell activation. Int J Biol Macromol 2024; 279:135394. [PMID: 39245093 DOI: 10.1016/j.ijbiomac.2024.135394] [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: 07/15/2024] [Revised: 08/30/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
The onset and progression of colorectal cancer is intricately linked to a multitude of factors. Among these, immune cells present within the tumor microenvironment play a pivotal role, particularly natural killer (NK) cells, which are essential for mediating anti-tumor immunity. This study aims to elucidate the mechanism by which the VWA2 protein facilitates the invasion and migration of colorectal cancer cells through the inhibition of NK cell activation. Understanding this molecular mechanism is crucial for deciphering the underlying processes involved in colorectal cancer. To achieve the study's objectives, various methodologies were employed, including cell culture techniques, transgenic technology, and assessments of NK cell functionality. The "limma" bioinformatics tool was utilised to identify differentially expressed genes (DEGs) between samples of colon cancer or polyps and normal tissue through transcriptome sequencing. Subsequent Wien analysis was conducted to pinpoint overlapping genes of interest. The impact of VWA2 on both the invasion and migration of colorectal cancer cell lines was assessed through experiments designed for the overexpression and knockout of VWA2.In addition, flow cytometry was employed to evaluate the activation status of NK cells, enabling an analysis of how VWA2 modulates relevant signaling pathways. The findings revealed that overexpression of VWA2 led to a marked inhibition of NK cell activation, which corresponded with reduced cytotoxic activity against tumor cells. Further examination indicated that VWA2 significantly amplified the migration and invasion capabilities of colorectal cancer cells by upregulating immunosuppressive factors while simultaneously downregulating pro-inflammatory factors. Conversely, the reduction of VWA2 expression was shown to markedly enhance NK cell functionality and decrease the invasive potential of colorectal cancer cells. Thus, the evidence suggests that the VWA2 protein actively promotes the migration and invasion of colorectal cancer cells primarily by suppressing NK cell activation, highlighting its potential role as a significant contributor to tumor progression in colorectal cancer.
Collapse
Affiliation(s)
- Yang Tian
- Department of Endoscope, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Qi Zhao
- Department of Endoscope, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Haowa Wu
- Department of Gastroenterology, Aviation General Hospital, Chaoyang District, Beijing 100012, China
| | - Jiaqi Guo
- Department of Gastroenterology, The Affiliated Hospital of Northwest University Xi'an No. 3 Hospital, Xi'an 710000, Shaanxi Province, China
| | - Huaxing Wu
- Department of Endoscope, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China.
| |
Collapse
|
5
|
Liu H, Lu Y, Zong J, Zhang B, Li X, Qi H, Yu T, Li Y. Engineering dendritic cell biomimetic membrane as a delivery system for tumor targeted therapy. J Nanobiotechnology 2024; 22:663. [PMID: 39465376 PMCID: PMC11520105 DOI: 10.1186/s12951-024-02913-7] [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: 05/17/2024] [Accepted: 10/07/2024] [Indexed: 10/29/2024] Open
Abstract
Targeted immunotherapies make substantial strides in clinical cancer care due to their ability to counteract the tumor's capacity to suppress immune responses. Advances in biomimetic technology with minimally immunogenic and highly targeted, are addressing issues of targeted drug delivery and disrupting the tumor's immunosuppressive environment to trigger immune activation. Specifically, the use of dendritic cell (DC) membranes to coat nanoparticles ensures targeted delivery due to DC's unique ability to activate naive T cells, spotlighting their role in immunotherapy aimed at disrupting the tumor microenvironment. The potential of DC's biomimetic membrane to mediate immune activation and target tumors is gaining momentum, enhancing the effectiveness of cancer treatments in conjunction with other immune responses. This review delves into the methodologies behind crafting DC membranes and the fusion of dendritic and tumor cell membranes for encapsulating therapeutic nanoparticles. It explores their applications and recent advancements in combating cancer, offering an all-encompassing perspective on DC biomimetic nanosystems, immunotherapy driven by antigen presentation, and the collaborative efforts of drug delivery in chemotherapy and photodynamic therapies. Current evidence shows promise in augmenting combined therapeutic approaches for cancer treatment and holds translational potential for various cancer treatments in a clinical setting.
Collapse
Affiliation(s)
- Huiyang Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, People's Republic of China
| | - Yiming Lu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, People's Republic of China
| | - Jinbao Zong
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China
| | - Bei Zhang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
| | - Xiaolu Li
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China
| | - Hongzhao Qi
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao, 266000, People's Republic of China.
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China.
| | - Yu Li
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, People's Republic of China.
| |
Collapse
|
6
|
Guo KC, Wang ZZ, Su XQ. Chinese Medicine in Colorectal Cancer Treatment: From Potential Targets and Mechanisms to Clinical Application. Chin J Integr Med 2024:10.1007/s11655-024-4115-8. [PMID: 39331211 DOI: 10.1007/s11655-024-4115-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2024] [Indexed: 09/28/2024]
Abstract
Colorectal cancer (CRC) is a global health challenge necessitating innovative therapeutic strategies. There is an increasing trend toward the clinical application of integrative Chinese medicine (CM) and Western medicine approaches. Chinese herbal monomers and formulations exert enhanced antitumor effects by modulating multiple signaling pathways in tumor cells, including inhibiting cell proliferation, inducing apoptosis, suppressing angiogenesis, reversing multidrug resistance, inhibiting metastasis, and regulating immunity. The synergistic effects of CM with chemotherapy, targeted therapy, immunotherapy, and nanovectors provide a comprehensive framework for CRC treatment. CM can mitigate drug toxicity, improve immune function, control tumor progression, alleviate clinical symptoms, and improve patients' survival and quality of life. This review summarizes the key mechanisms and therapeutic strategies of CM in CRC, highlighting its clinical significance. The potential for CM and combination with conventional treatment modalities is emphasized, providing valuable insights for future research and clinical practice.
Collapse
Affiliation(s)
- Ke-Chen Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Zao-Zao Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiang-Qian Su
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
7
|
Janduang S, Cotchim S, Kongkaew S, Srilikhit A, Wannapob R, Kanatharana P, Thavarungkul P, Limbut W. Synthesis of flower-like ZnO nanoparticles for label-free point of care detection of carcinoembryonic antigen. Talanta 2024; 277:126330. [PMID: 38833905 DOI: 10.1016/j.talanta.2024.126330] [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: 09/17/2023] [Revised: 05/21/2024] [Accepted: 05/26/2024] [Indexed: 06/06/2024]
Abstract
In this work, flower-like ZnO nanoparticles (ZnONPs) were synthesized using zinc nitrate (Zn(NO3)2 6H2O) as a precursor with KOH. The morphology of the ZnONPs was controlled by varying the synthesis temperature at 50, 75 and 95 °C. The morphology and structure of ZnONPs were characterized using Scanning Electron Microscopy, and X-Ray Diffraction and Brunauer-Emmett Teller analysis. ZnONPs were successfully synthesized by a simple chemical precipitation method. A synthesis temperature of 75 °C produced the most suitable flower-like ZnONPs, which were combined with graphene nanoplatelets to develop a label-free electrochemical immunosensor for the detection of the colon cancer biomarker carcinoembryonic antigen in human serum. Under optimum conditions, the developed immunosensor showed a linear range of 0.5-10.0 ng mL-1 with a limit of detection of 0.44 ng mL-1. The label-free electrochemical immunosensor exhibited good selectivity, reproducibility, and repeatability, and recoveries were excellent. The immunosensor is used with a Near-Field Communication potentiostat connected to a smartphone to facilitate point-of-care cancer detection in low-resource locations.
Collapse
Affiliation(s)
- Santipap Janduang
- Center of Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Division of Physical Science, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Suparat Cotchim
- Center of Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Supatinee Kongkaew
- Center of Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Angkana Srilikhit
- Center of Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Division of Physical Science, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Rodtichoti Wannapob
- Silicon Craft Technology PLC, No. 40, Thetsabanrangsannua Rd., Ladyao, Chatuchak, Bangkok, 10900, Thailand
| | - Proespichaya Kanatharana
- Center of Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Division of Physical Science, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Panote Thavarungkul
- Center of Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Division of Physical Science, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Warakorn Limbut
- Center of Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand; Forensic Science Innovation and Service Center, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand.
| |
Collapse
|
8
|
Hu Y, Wu X, Tan X, Zhang J. Hsa_circRNA_007630 knockdown delays colon cancer progression by modulation of ferroptosis via miR-506-3p/AURKA axis. J Biochem Mol Toxicol 2024; 38:e23771. [PMID: 39015057 DOI: 10.1002/jbt.23771] [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: 02/04/2024] [Revised: 06/06/2024] [Accepted: 07/05/2024] [Indexed: 07/18/2024]
Abstract
Colon cancer contributes to high mortality rates internationally that has seriously endangered human health. Aurora kinase A (AURKA) served as a key molecule in colon cancer. However, its role of AURKA on regulating ferroptosis in colon cancer and their possible interactions with miRNAs and circRNAs remain still elusive. Comprehensive bioinformatics analysis after RNA-sequencing was conducted to determine the differentially expressed genes (DEGs), ferroptosis-related DEGs and hub genes. The direct relationship between miR-506-3p and hsa_circRNA_007630 or AURKA was predicted, then verified by dual luciferase reporter and quantitative real-time polymerase chain reaction. The rescue experiments were conducted by cotransfection with si-hsa_circRNA_007630, miR-506-3p inhibitor or pcDNA-AURKA in HT29 cells. Erastin was used to induce ferroptosis in HT29 cells and validated by detecting levels of intracellular Fe2+, lipid reactive oxygen species, glutathione, malondialdehyde and ferroptosis markers expression. We screened a total of 331 DEGs, 26 ferroptosis-related genes, among which 3 hub genes were identified through PPI network analysis. Therein, AURKA expression was elevated in colon cancer cells. Moreover, AURKA was targeted by miR-506-3p, and hsa_circRNA_007630 operated as miR-506-3p sponge. The effect of hsa_circRNA_007630 depletion on the inhibiting malignant phenotypes of HT29 cells was rescued by inhibition of miR-506-3p or AURKA overexpression. Additionally, AURKA reduced erastin-induced ferroptosis in HT29 cells. Depletion of circRNA_007630 exerts as a suppressive role in colon cancer through a novel miR-506-3p/AURKA pathway related to ferroptosis, and might become a novel marker for colon cancer.
Collapse
Affiliation(s)
- Ying Hu
- Department of Gastroenterology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, China
| | - Xiongjian Wu
- Department of Gastroenterology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, China
| | - Xiaobin Tan
- Department of Clinical Laboratory, First Affiliated Hospital of Gannan Medical University, Ganzhou City, China
| | - Jingzhi Zhang
- Department of Gastroenterology, Ganzhou People's Hospital (Ganzhou Hospital Affiliated to Nanchang University), Ganzhou City, China
| |
Collapse
|
9
|
Gui Y, Deng X, Li N, Zhao L. PRELP reduce cell stiffness and adhesion to promote the growth and metastasis of colorectal cancer cells by binding to integrin α5. Exp Cell Res 2024; 441:114151. [PMID: 38992455 DOI: 10.1016/j.yexcr.2024.114151] [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: 12/14/2023] [Revised: 06/19/2024] [Accepted: 07/03/2024] [Indexed: 07/13/2024]
Abstract
PRELP is thought to be an inhibitor of the development and progression of a variety of malignancies. Metastasis is a major cause of death in patients with colorectal cancer, but the mechanism underlying the role of PRELP in colorectal cancer metastasis remains poorly understood. In this study, we found that PRELP was significantly higher in metastatic tissues than in non-metastatic tissues of colorectal cancer and was closely associated with poor prognosis of colorectal cancer patients. PRELP promotes growth and metastasis of colorectal cancer cells. PRELP reduces cell stiffness and adhesion. PRELP promoted EMT in colorectal cancer cells and that PRELP bind to integrin α5 to activate the integrin α5/FAK/AKT signaling pathway. In conclusion, we demonstrate that PRELP is upregulated in metastatic colorectal cancer, providing a potential prognostic marker and therapeutic target for the clinical management of metastatic colorectal cancer from a biomechanical perspective.
Collapse
Affiliation(s)
- Yajun Gui
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 41001l, China; Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China
| | - Xiangying Deng
- Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Namei Li
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 41001l, China; Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China
| | - Lin Zhao
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 41001l, China; Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China.
| |
Collapse
|
10
|
Andoh V, Ocansey DKW, Naveed H, Wang N, Chen L, Chen K, Mao F. The Advancing Role of Nanocomposites in Cancer Diagnosis and Treatment. Int J Nanomedicine 2024; 19:6099-6126. [PMID: 38911500 PMCID: PMC11194004 DOI: 10.2147/ijn.s471360] [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: 03/29/2024] [Accepted: 06/12/2024] [Indexed: 06/25/2024] Open
Abstract
The relentless pursuit of effective cancer diagnosis and treatment strategies has led to the rapidly expanding field of nanotechnology, with a specific focus on nanocomposites. Nanocomposites, a combination of nanomaterials with diverse properties, have emerged as versatile tools in oncology, offering multifunctional platforms for targeted delivery, imaging, and therapeutic interventions. Nanocomposites exhibit great potential for early detection and accurate imaging in cancer diagnosis. Integrating various imaging modalities, such as magnetic resonance imaging (MRI), computed tomography (CT), and fluorescence imaging, into nanocomposites enables the development of contrast agents with enhanced sensitivity and specificity. Moreover, functionalizing nanocomposites with targeting ligands ensures selective accumulation in tumor tissues, facilitating precise imaging and diagnostic accuracy. On the therapeutic front, nanocomposites have revolutionized cancer treatment by overcoming traditional challenges associated with drug delivery. The controlled release of therapeutic agents from nanocomposite carriers enhances drug bioavailability, reduces systemic toxicity, and improves overall treatment efficacy. Additionally, the integration of stimuli-responsive components within nanocomposites enables site-specific drug release triggered by the unique microenvironment of the tumor. Despite the remarkable progress in the field, challenges such as biocompatibility, scalability, and long-term safety profiles remain. This article provides a comprehensive overview of recent developments, challenges, and prospects, emphasizing the transformative potential of nanocomposites in revolutionizing the landscape of cancer diagnostics and therapeutics. In Conclusion, integrating nanocomposites in cancer diagnosis and treatment heralds a new era for precision medicine.
Collapse
Affiliation(s)
- Vivian Andoh
- School of Life Sciences, Jiangsu University, Zhenjiang, People’s Republic of China
| | - Dickson Kofi Wiredu Ocansey
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu, People’s Republic of China
- Directorate of University Health Services, University of Cape Coast, Cape Coast, Central Region, CC0959347, Ghana
| | - Hassan Naveed
- School of Life Sciences, Jiangsu University, Zhenjiang, People’s Republic of China
| | - Naijian Wang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Liang Chen
- School of Life Sciences, Jiangsu University, Zhenjiang, People’s Republic of China
| | - Keping Chen
- School of Life Sciences, Jiangsu University, Zhenjiang, People’s Republic of China
| | - Fei Mao
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu, People’s Republic of China
| |
Collapse
|
11
|
Elattar KM, Al-Otibi FO, El-Hersh MS, Attia AA, Eldadamony NM, Elsayed A, Menaa F, Saber WI. Multifaceted chemical and bioactive features of Ag@TiO 2 and Ag@SeO 2 core/shell nanoparticles biosynthesized using Beta vulgaris L. extract. Heliyon 2024; 10:e28359. [PMID: 38560145 PMCID: PMC10979172 DOI: 10.1016/j.heliyon.2024.e28359] [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: 12/15/2023] [Revised: 02/25/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Due to increasing concerns about environmental impact and toxicity, developing green and sustainable methods for nanoparticle synthesis is attracting significant interest. This work reports the successful green synthesis of silver (Ag), silver-titanium dioxide (Ag@TiO2), and silver-selenium dioxide (Ag@SeO2) nanoparticles (NPs) using Beta vulgaris L. extract. Characterization by XRD, SEM, TEM, and EDX confirmed the successful formation of uniformly distributed spherical NPs with controlled size (25 ± 4.9 nm) and desired elemental composition. All synthesized NPs and the B. vulgaris extract exhibited potent free radical scavenging activity, indicating significant antioxidant potential. However, Ag@SeO2 displayed lower hemocompatibility compared to other NPs, while Ag@SeO2 and the extract demonstrated reduced inflammation in a carrageenan-induced paw edema animal model. Interestingly, Ag@TiO2 and Ag@SeO2 exhibited strong antifungal activity against Rhizoctonia solani and Sclerotia sclerotium, as evidenced by TEM and FTIR analyses. Generally, the findings suggest that B. vulgaris-derived NPs possess diverse biological activities with potential applications in various fields such as medicine and agriculture. Ag@TiO2 and Ag@SeO2, in particular, warrant further investigation for their potential as novel bioactive agents.
Collapse
Affiliation(s)
- Khaled M. Elattar
- Unit of Genetic Engineering and Biotechnology, Faculty of Science, Mansoura University, El-Gomhoria Street, Mansoura, 35516, Egypt
| | - Fatimah O. Al-Otibi
- Botany and Microbiology Department, Faculty of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Mohammed S. El-Hersh
- Microbial Activity Unit, Department of Microbiology, Soils, Water and Environment Research Institute, Agricultural Research Center, Giza, 12619, Egypt
| | - Attia A. Attia
- Department of Botany and Microbiology, Faculty of Science, Benha University, Benha, Egypt
| | - Noha M. Eldadamony
- Seed Pathology Department, Plant Pathology Research Institute, Agricultural Research Center, Giza, 12619, Egypt
| | - Ashraf Elsayed
- Botany Department, Faculty of Science, Mansoura University, Elgomhouria St., Mansoura, 35516, Egypt
| | - Farid Menaa
- Department of Biomedical and Environmental Engineering (BEE), Fluorotronics, Inc. California Innovation Corporation, San Diego, CA 92037, USA
| | - WesamEldin I.A. Saber
- Microbial Activity Unit, Department of Microbiology, Soils, Water and Environment Research Institute, Agricultural Research Center, Giza, 12619, Egypt
| |
Collapse
|
12
|
Sedky NK, Fawzy IM, Hassan A, Mahdy NK, Attia RT, Shamma SN, Alfaifi MY, Elbehairi SE, Mokhtar FA, Fahmy SA. Innovative microwave-assisted biosynthesis of copper oxide nanoparticles loaded with platinum(ii) based complex for halting colon cancer: cellular, molecular, and computational investigations. RSC Adv 2024; 14:4005-4024. [PMID: 38288146 PMCID: PMC10823359 DOI: 10.1039/d3ra08779d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 01/21/2024] [Indexed: 01/31/2024] Open
Abstract
In the current study, we biosynthesized copper oxide NPs (CuO NPs) utilizing the essential oils extracted from Boswellia carterii oleogum resin, which served as a bioreductant and capping agent with the help of microwave energy. Afterwards, the platinum(ii) based anticancer drug, carboplatin (Cr), was loaded onto the CuO NPs, exploiting the electrostatic interactions forming Cr@CuO NPs. The produced biogenic NPs were then characterized using zeta potential (ZP), high-resolution transmission electron microscopy (HRTEM), X-ray diffraction spectroscopy (XRD), and Fourier transform infrared spectroscopy (FTIR) techniques. In addition, the entrapment efficiency and release profile of the loaded Cr were evaluated. Thereafter, SRB assay was performed, where Cr@CuO NPs demonstrated the highest cytotoxic activity against human colon cancer cells (HCT-116) with an IC50 of 5.17 μg mL-1, which was about 1.6 and 2.2 folds more than that of Cr and CuO NPs. Moreover, the greenly synthesized nanoparticles (Cr@CuO NPs) displayed a satisfactory selectivity index (SI = 6.82), which was far better than the free Cr treatment (SI = 2.23). Regarding the apoptosis assay, the advent of Cr@CuO NPs resulted in an immense increase in the cellular population percentage of HCT-116 cells undergoing both early (16.02%) and late apoptosis (35.66%), significantly surpassing free Cr and CuO NPs. A study of HCT-116 cell cycle kinetics revealed the powerful ability of Cr@CuO NPs to trap cells in the Sub-G1 and G2 phases and impede the G2/M transition. RT-qPCR was utilized for molecular investigations of the pro-apoptotic (Bax and p53) and antiapoptotic genes (Bcl-2). The novel Cr@CuO NPs treatment rose above single Cr or CuO NPs therapy in stimulating the p53-Bax mediated mitochondrial apoptosis. The cellular and molecular biology investigations presented substantial proof of the potentiated anticancer activity of Cr@CuO NPs and the extra benefits that could be obtained from their use.
Collapse
Affiliation(s)
- Nada K Sedky
- Department of Biochemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Administrative Capital Cairo Egypt
| | - Iten M Fawzy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Future University in Egypt Cairo 11835 Egypt
| | - Afnan Hassan
- Biomedical Sciences Program, Zewail City of Science and Technology Giza 12578 Egypt
| | - Noha Khalil Mahdy
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University Kasr El-Aini Street 11562 Cairo Egypt
| | - Reem T Attia
- Department of Pharmacology and Toxicology and Biochemistry, Faculty of Pharmacy, Future University in Egypt Cairo 11835 Egypt
| | - Samir N Shamma
- Institute of Global Health and Human Ecology, School of Sciences & Engineering, The American University in Cairo AUC Avenue, P.O. Box 74 New Cairo 11835 Egypt
| | - Mohammad Y Alfaifi
- King Khalid University, Faculty of Science, Biology Department Abha 9004 Saudi Arabia
| | - Serag Eldin Elbehairi
- King Khalid University, Faculty of Science, Biology Department Abha 9004 Saudi Arabia
| | - Fatma A Mokhtar
- Department of Pharmacognosy, Faculty of Pharmacy, El Saleheya El Gadida University El Saleheya El Gadida Sharkia 44813 Egypt
| | - Sherif Ashraf Fahmy
- Department of Chemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Capital Cairo 11835 Egypt +20 1222613344
| |
Collapse
|
13
|
Guo Z, Song H, Tian Y, Xu J, Zhang G, Guo Y, Shen R, Wang D. SiRNF8 Delivered by DNA Framework Nucleic Acid Effectively Sensitizes Chemotherapy in Colon Cancer. Int J Nanomedicine 2024; 19:171-188. [PMID: 38204601 PMCID: PMC10777867 DOI: 10.2147/ijn.s437859] [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: 08/30/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Background The evident side effects and decreased drug sensitivity significantly restrict the use of chemotherapy. However, nanoparticles based on biomaterials are anticipated to address this challenge. Methods Through bioinformatics analysis and colon cancer samples, we initially investigated the expression level of RNF8 in colon cancer. Next, we constructed nanocarrier for delivering siRNF8 based on DNA tetrahedron (si-Tet), and Doxorubicin (DOX) was further intercalated into the DNA structure (si-DOX-Tet) for combination therapy. Further, the effects and mechanism of RNF8 inhibition on the sensitivity of colon cancer cells to DOX chemotherapy have also been studied. Results RNF8 expression was increased in colon cancer. Agarose gel electrophoresis, transmission electron microscopy, and size distribution and potential analysis confirmed the successful preparation of the two nanoparticles, with particle sizes of 10.29 and 37.29 nm, respectively. Fluorescence imaging reveals that the carriers can be internalized into colon cancer cells and escape from lysosomes after 12 hours of treatment, effectively delivering siRNF8 and DOX. Importantly, Western blot analysis verified treatment with 50nM si-Tet silenced RNF8 expression by approximately 50% in colon cancer cells, and combined treatment significantly inhibited cell proliferation. Furthermore, the CCK-8 assay demonstrated that si-Tet treatment enhanced the sensitivity of colon cancer cells to the three chemotherapeutic drugs. Significant more DNA damage was detected after treatment with both si-Tet or si-DOX-Tet. Further flow cytometry analysis revealed that si-DOX-Tet treatment led to significantly more apoptosis, approximately 1.6-fold higher than treatment with DOX alone. Mechanistically, inhibiting RNF8 led to decreased ABCG2 expression and DOX efflux, but increased DNA damage, thereby enhancing the chemotherapeutic effect of DOX. Conclusion We have successfully constructed si-DOX-Tet. By inhibiting the expression of RNF8, it enhances the chemotherapy sensitivity of DOX. Therefore, this tetrahedral FNA nanocarrier offers a new approach for the combined treatment of colon cancer.
Collapse
Affiliation(s)
- Zhao Guo
- Department of Anatomy and Histology, Lanzhou University School of Basic Medical Sciences, Lanzhou, 730000, People’s Republic of China
| | - Haoyun Song
- Department of Anatomy and Histology, Lanzhou University School of Basic Medical Sciences, Lanzhou, 730000, People’s Republic of China
| | - Yingxia Tian
- Department of Internal Medicine, Gansu Provincial Academic Institute for Medical Research, Lanzhou, 730050, People’s Republic of China
| | - Jie Xu
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, Gansu, 730030, People’s Republic of China
| | - Guokun Zhang
- Department of Anatomy and Histology, Lanzhou University School of Basic Medical Sciences, Lanzhou, 730000, People’s Republic of China
| | - Yanan Guo
- Department of Anatomy and Histology, Lanzhou University School of Basic Medical Sciences, Lanzhou, 730000, People’s Republic of China
| | - Rong Shen
- Department of Anatomy and Histology, Lanzhou University School of Basic Medical Sciences, Lanzhou, 730000, People’s Republic of China
| | - Degui Wang
- Department of Anatomy and Histology, Lanzhou University School of Basic Medical Sciences, Lanzhou, 730000, People’s Republic of China
| |
Collapse
|
14
|
Du J. Study of Therapeutic Mechanisms of Bupi Yichang Formula against Colon Cancer Based on Network Pharmacology, Machine Learning, and Experimental Verification. Crit Rev Immunol 2024; 44:67-87. [PMID: 38421706 DOI: 10.1615/critrevimmunol.2023051509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Bupi Yichang formula (BPYCF) has shown the anti-cancer potential; however, its effects on colon cancer and the mechanisms remain unknown. This study intended to explore the effects of BPYC on colon cancer and its underlying mechanisms. BPYCF-related and colon cancer-related targets were acquired from public databases, followed by differentially expressed genes (DEG) identification. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed using clusterProfiler. A protein-protein interaction (PPI) network was constructed using STRING database. CytoHubba and MCODE to screen the hub targets. A diagnostic model was built using random forest algorithm. Molecular docking was conducted using PyMOL and AutoDock. High-performance liquid chromatograph-mass spectrometry (HPLC-MS) analysis and in vitro validation were performed. Forty-six overlapping targets of BPYCF-related, colon cancer-related targets, and DEGs were obtained. GO and KEGG analyses showed that the targets were mainly enriched in response to lipopolysaccharide, neuronal cell body, protein serine/threonine/tyrosine, as well as C-type lectin receptor, NOD-like receptor, and TNF signaling pathways. Five targets were identified as the pivotal targets, among which, NOS3, CASP8, RIPK3, and TNFRSF10B were stably docked with the core active component, naringenin. Naringenin was also identified from the BPYCF sample through HPLC-MS analysis. In vitro experiments showed that BPYCF inhibited cell viability, reduced NOS3 expression, and elevated CASP8, RIPK3, and TNFRSF10B expression in colon cancer cells. BPYCF might treat colon cancer mainly by regulating NOS3, CASP8, RIPK3, and TN-FRSF10B. This study first revealed the therapeutic effects and mechanisms of BPYCF against colon cancer, paving the path for the development of targeted therapeutic strategies for this cancer in the clinic.
Collapse
Affiliation(s)
- Juan Du
- Beijing Friendship Hospital, Capital Medical University
| |
Collapse
|
15
|
Kong Y, Xu L, Cao J. Preparation of a Novel Multifunctional Cationic Liposome Drug-carrying System and its Functional Study on Lung Cancer. Anticancer Agents Med Chem 2024; 24:1085-1095. [PMID: 38803174 DOI: 10.2174/0118715206294695240522075454] [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: 01/24/2024] [Revised: 03/16/2024] [Accepted: 04/09/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Low-dose chemotherapy is a promising treatment strategy that may be improved by controlled delivery. OBJECTIVE This study aimed to design polyethylene glycol-stabilized bilayer-decorated magnetic Cationic Liposomes (CLs) as a drug delivery system for integrated functional studies of lung cancer cell therapy and imaging. METHODS A novel multifunctional folic acid targeting magnetic CLs docetaxel drug-loading system (FA-CLs-Fe- DOC) was prepared and tested for its physical properties, encapsulation rate and drug release performance. The feasibility of FA-CLs-Fe-DOC ability to inhibit tumor cells and act as an MRI contrast agent was investigated in vitro, and the target recognition and therapeutic ability of FA-CLs-Fe-DOC was studied in vivo. RESULTS FA-CLs-Fe-DOC had a particle size of 221.54 ± 6.42 nm and a potential of 28.64 ± 3.56 mv, with superparamagnetic properties and better stability. The encapsulation rate was 95.36 ± 1.63%, and the drug loading capacity was 9.52 ± 0.22%, which possessed the drug slow-release performance and low cytotoxicity and could effectively inhibit the proliferation of lung cancer cells, promoting apoptosis of lung cancer cells. MRI showed that it had the function of tracking and localization of lung cancer cells. In vivo experiments confirmed the targeted recognition property and therapeutic function of lung cancer cells. CONCLUSION In this study, we successfully prepared an FA-CLs-Fe-DOC capable of specifically targeting lung cancer cells with integrated functions of efficient lung cancer cell killing and imaging localization. This targeted drug packaging technology may provide a new strategy for the design of integrated carriers for targeted cancer therapy and imaging.
Collapse
Affiliation(s)
- Yi Kong
- The Second Department of Thoracic Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410013, Hunan Province, P.R. China
| | - Li Xu
- The Second Department of Thoracic Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha 410013, Hunan Province, P.R. China
| | - Jun Cao
- The First Department of Thoracic Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, Hunan Province, P.R. China
| |
Collapse
|
16
|
Sun B, Yue SG. Expression of long noncoding RNA MEG3 and microRNA-302b-3p in colon cancer: Correlation with clinical stage and value in predicing prognosis after surgical treatment. Shijie Huaren Xiaohua Zazhi 2023; 31:981-988. [DOI: 10.11569/wcjd.v31.i23.981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/08/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND More and more long non-coding RNAs and microRNAs have been found to have significant changes in expression levels during the occurrence and development of tumors, which can affect the expression of tumor suppressor genes or oncogenes and play an important role in the proliferation and metastasis of cancer cells.
AIM To investigate the correlation between the expression of long non-coding RNA maternal imprinted gene 3 (LncRNA MEG3) and microRNA (miR)-302b-3p in colon cancer and clinical stage and analyze their value in predicting the prognosis after surgical treatment.
METHODS A total of 97 patients with colon cancer treated at Jinhua Hospital of TCM from January 2017 to March 2022 were selected to compare the expression of LncRNA MEG3 and miR-302b-3p in different tissues, analyze the correlation between the expression of LncRNA MEG3 and miR-302b-3p and clinical pathological characteristics, compare the recurrence in patients with different LncRNA MEG3 and miR-302b-3p expression, analyze the factors affecting the recurrence of colon cancer after surgery, and analyze the impact of the interaction between LncRNA MEG3 and miR-302b-3p on the recurrence of colon cancer. The predictive value of LncRNA MEG3 and miR-302b-3p expression for the recurrence of colon cancer after surgery was evaluated.
RESULTS The expression of LncRNA MEG3 and miR-302b-3p in colon cancer tissues was lower than that in tumor-adjacent tissues (P < 0.05). The expression of LncRNA MEG3 and miR-302b-3p in colon cancer tissues was not correlated with sex, age, or tumor size (P > 0.05), but was correlated with tumor differentiation, clinical stage, and lymph node metastasis (P < 0.05). In colon cancer tissues, the recurrence rate in patients with high LncRNA MEG3 and miR-302b-3p expression was lower than that of patients with lower LncRNA MEG3 and miR-302b-3p expression (P < 0.05). Tumor differentiation degree, clinical stage, and lymph node metastasis were all identified to be risk factors for colon cancer recurrence, and LncRNA MEG3 and miR-302b-3p expression were protective factors for colon cancer recurrence (P < 0.05). The interaction analysis showed that the synergistic effect of simultaneous exposure to LncRNA MEG3 and miR-302b-3p was 15.888 times greater than the effect of exposure to either LncRNA MEG3 or miR-302b-3p alone, and when simultaneously exposing to both, 56.98% of the risk of colon cancer recurrence was attributed to their synergistic effect. The area under the curve (AUC) (95% confidence interval [CI]) of LncRNA MEG3 and miR-302b-3p in predicting the prognosis of colon cancer patients was 0.720 (0.620-0.807) and 0.767 (0.670-0.847), respectively, and that of the combined prediction was 0.892 (0.813-0.946), with a sensitivity and specificity of 92.31% and 83.33%, respectively, which were significantly higher than those of either LncRNA MEG3 or miR-302b-3p alone.
CONCLUSION The down-regulated expression of LncRNA MEG3 and miR-302b-3p in colon cancer is related to clinical stage. Clinical detection of their expression can be used to determine the malignant degree of tumor and predict the prognosis of surgical treatment, thus providing reference for adjustment of clinical treatment plan.
Collapse
Affiliation(s)
- Bing Sun
- Second Department of Surgery, Jinhua Hospital of Traditional Chinese Medicine, Jinhua 321017, Zhejiang Province, China
| | - Shi-Guo Yue
- Second Department of Surgery, Jinhua Hospital of Traditional Chinese Medicine, Jinhua 321017, Zhejiang Province, China
| |
Collapse
|
17
|
Ding Y, Zhao H, Niu W, Zhang J, Zheng X, Liu Y, Zhang J, Li C, Yu B. M2 Macrophage-Derived Extracellular Vesicles Containing MicroRNA-501-3p Promote Colon Cancer Progression Through the SETD7/DNMT1/SOCS3 Axis. Dis Colon Rectum 2023; 66:e1234-e1245. [PMID: 37695661 DOI: 10.1097/dcr.0000000000002986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
BACKGROUND Macrophage-derived extracellular vesicles with microRNAs can cause and develop colon cancer. OBJECTIVE To investigate M2 macrophage-derived extracellular vesicles and colon cancer. DESIGN A prospective and experimental study of M2 macrophage-derived extracellular vesicles in colon cancer. SETTING This study was completed at the Fourth Hospital of Hebei Medical University. PATIENTS Patients with colon cancer who had undergone surgical resection. MAIN OUTCOME MEASURES Suppressor of cytokine signaling 3, miR-501-3p, SET domain containing 7, and DNA methyltransferase 1 were measured in colon cancer samples. Multiple experiments determined suppressor of cytokine signaling 3, miR-501-3p, SET domain containing 7, and DNA methyltransferase 1 binding affinity. M2 macrophages were cultivated from M0 macrophages isolated from peripheral blood mononuclear cells of a healthy donor and polarized to produce extracellular vesicles. Gain- or loss-of-function tests using colon cancer cells and M2 macrophage-derived extracellular vesicles revealed cell biological processes. Finally, animal models were created to test how miR-501-3p from M2-extracellular vesicles affects tumor growth via the SET domain containing 7/DNA methyltransferase 1/suppressor of cytokine signaling 3. RESULTS Colon cancer increased miR-501-3p and DNA methyltransferase 1 and downregulated suppressor of cytokine signaling 3 and SET domain containing 7. miR-151-3p inhibited SET domain containing 7, upregulating DNA methyltransferase 1. Increased promoter methylation by DNA methyltransferase 1 decreased suppressor of cytokine signaling 3 expression. M2-EVs with miR-501-3p regulated the SET domain containing 7/DNA methyltransferase 1/suppressor of cytokine signaling 3 axis to induce apoptosis and colon cancer cell growth, invasion, and migration. M2-EV-delivered miR-501-3p also regulated the SET domain containing 7/DNA methyltransferase 1/suppressor of cytokine signaling 3 axis to promote tumor growth in animals. LIMITATIONS Further research is needed in clinical application of M2 macrophage-derived extracellular vesicles containing miR-501-3p as a biomarker of colon cancer. CONCLUSIONS M2 macrophage-derived extracellular vesicles with miR-501-3p regulate the SET domain containing 7/DNA methyltransferase 1/suppressor of cytokine signaling 3 axis to promote colon cancer. LAS VESCULAS EXTRACELULARES DERIVADAS DE MACRFAGOS M QUE CONTIENEN MICROARNP PROMUEVEN LA PROGRESIN DEL CNCER DE COLON A TRAVS DEL EJE SETD/DNMT/SOCS ANTECEDENTES:Las vesículas extracelulares derivadas de macrófagos con microARN pueden causar y desarrollar cáncer de colon.OBJETIVO:Investigamos las vesículas extracelulares derivadas de macrófagos M2 y el cáncer de colon.DISEÑO:Un estudio prospectivo y experimental de vesículas extracelulares derivadas de macrófagos M2 en el cáncer de colon.ESCENARIO:Este estudio se completó en el Cuarto Hospital de la Universidad Médica de Hebei.PACIENTES:Pacientes con cáncer de colon sometidos a resección quirúrgica.PRINCIPALES MEDIDAS DE RESULTADO:Se midieron el supresor de la señalización de citoquinas 3, miR-501-3p, SETD7 y la ADN metiltransferasa 1 en muestras de cáncer de colon. Múltiples experimentos determinaron la afinidad de unión del supresor de la señalización de citoquinas 3, de miR-501-3p, de SETD7 y de la ADN metiltransferasa 1. Los macrófagos M2 se cultivaron a partir de macrófagos M0 aislados de células mononucleares de sangre periférica de donantes sanos y se polarizaron para producir vesículas extracelulares. Las pruebas de ganancia o pérdida de función utilizando células de cáncer de colon y vesículas extracelulares derivadas de macrófagos M2 revelaron procesos biológicos celulares. Finalmente, se crearon modelos animales para probar cómo miR-501-3p de vesículas extracelulares M2 afecta el crecimiento tumoral a través del SETD7/ADN metiltransferasa 1/supresor de la señalización de citocinas 3.RESULTADOS:El cáncer de colon aumentó el miR-501-3p y la ADN metiltransferasa 1 y reguló negativamente el supresor de la señalización de citoquinas 3 y SETD7. miR-151-3p inhibió SETD7, regulando positivamente la ADN metiltransferasa 1. El aumento de la metilación del promotor por la ADN metiltransferasa 1 produjo disminución de la expresión del supresor de señalización de citocinas 3. Los M2-EV con miR-501-3p regularon el eje SETD7/ADN metiltransferasa 1/supresor de la señalización de citocinas 3 para inducir apoptosis y crecimiento, invasión y migración de células de cáncer de colon. El miR-501-3p administrado por M2-EV también reguló el eje SETD7/ADN metiltransferasa 1/supresor de la señalización de citocinas 3 para promover el crecimiento tumoral en animales.LIMITACIONES:Se necesita más investigación en la aplicación clínica de vesículas extracelulares derivadas de macrófagos M2 que contienen miR-501-3p como biomarcador de cáncer de colon.CONCLUSIONES:Las vesículas extracelulares derivadas de macrófagos M2 con miR-501-3p regulan el eje SETD7/ADN metiltransferasa 1/supresor de la señalización de citocinas 3 para promover el cáncer de colon. (Traducción-Dr. Felipe Bellolio ).
Collapse
Affiliation(s)
- Yuanyi Ding
- The Second Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Huijin Zhao
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Wenbo Niu
- The Second Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Juan Zhang
- The Second Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Xiaochuan Zheng
- The Second Department of General Surgery, Fengning Manchu Autonomous County Hospital, Chengde, People's Republic of China
| | - Youqiang Liu
- The Second Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Jianfeng Zhang
- The Second Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Chenhui Li
- The Second Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Bin Yu
- The Second Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| |
Collapse
|
18
|
Li S, Pei H, He S, Liang H, Guo R, Liu N, Mo Z. Chiral Carbon Dots and Chiral Carbon Dots with Circularly Polarized Luminescence: Synthesis, Mechanistic Investigation and Applications. Chem Asian J 2023; 18:e202300770. [PMID: 37819766 DOI: 10.1002/asia.202300770] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/07/2023] [Accepted: 10/10/2023] [Indexed: 10/13/2023]
Abstract
Chiral carbon dots (CCDs) can be widely used in various fields such as chiral recognition, chiral catalysis and biomedicine because of their unique optical properties, low toxicity and good biocompatibility. In addition, CCDs with circularly polarized luminescence (CPL) can be synthesized, thus broadening the prospects of CCDs applications. Since the research on CCDs is still in its infancy, this paper reviews the chiral origin, formation mechanism, chiral evolution, synthesis and emerging applications of CCDs, with a special focus on CCDs with CPL activity. It is hoped that it will provide some reference to solve the current problems faced by CCDs. Finally, the opportunities and challenges of the current research on CCDs are described, and their future development trends have also been prospected.
Collapse
Affiliation(s)
- Shijing Li
- Research Center of Gansu Military and Civilian Integration Advanced Structural Materials, Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Hebing Pei
- Research Center of Gansu Military and Civilian Integration Advanced Structural Materials, Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Simin He
- Research Center of Gansu Military and Civilian Integration Advanced Structural Materials, Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Hao Liang
- Research Center of Gansu Military and Civilian Integration Advanced Structural Materials, Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Ruibin Guo
- Research Center of Gansu Military and Civilian Integration Advanced Structural Materials, Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Nijuan Liu
- Research Center of Gansu Military and Civilian Integration Advanced Structural Materials, Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Zunli Mo
- Research Center of Gansu Military and Civilian Integration Advanced Structural Materials, Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| |
Collapse
|
19
|
Alyami MH, Musallam AA, Ibrahim TM, Mahdy MA, Elnahas HM, Aldeeb RA. The Exploitation of pH-Responsive Eudragit-Coated Mesoporous Silica Nanostructures in the Repurposing of Terbinafine Hydrochloride for Targeted Colon Cancer Inhibition: Design Optimization, In Vitro Characterization, and Cytotoxicity Assessment. Pharmaceutics 2023; 15:2677. [PMID: 38140018 PMCID: PMC10747614 DOI: 10.3390/pharmaceutics15122677] [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: 10/24/2023] [Revised: 11/13/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Targeted drug delivery is achieving great success in cancer therapy due to its potential to deliver drugs directly to the action site. Terbinafine hydrochloride (TER) is a broad-spectrum anti-fungal drug that has been found to have some potential anti-tumor effects in the treatment of colon cancer. We aimed here to design and develop pH-sensitive Eudragit (Eud)-coated mesoporous silica nanostructures (MSNs) to control drug release in response to changes in pH. The diffusion-supported loading (DiSupLo) technique was applied for loading TER into the MSNs. The formulation was optimized by a D-optimal design, which permits the concurrent assessment of the influence of drug/MSN%, coat concentration, and MSN type on the drug entrapment efficiency (EE) and its release performance. The optimal formula displayed a high EE of 96.49%, minimizing the release in pH 1.2 to 16.15% and maximizing the release in pH 7.4 to 78.09%. The cytotoxicity of the optimal formula on the colon cancer cells HT-29 was higher than it was with TER alone by 2.8-fold. Apoptosis in cancer cells exposed to the optimum formula was boosted as compared to what it was with the plain TER by 1.2-fold and it was more efficient in arresting cells during the G0/G1 and S stages of the cell cycle. Accordingly, the repurposing of TER utilizing Eud/MSNs is a promising technique for targeted colon cancer therapy.
Collapse
Affiliation(s)
- Mohammad H. Alyami
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 66462, Saudi Arabia
| | - Abeer A. Musallam
- Department of Pharmaceutics, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City 12582, Egypt
| | - Tarek M. Ibrahim
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Mahmoud A. Mahdy
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Hanan M. Elnahas
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Reem A. Aldeeb
- Department of Pharmaceutics, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City 12582, Egypt
| |
Collapse
|
20
|
Gowda BHJ, Ahmed MG, Alshehri SA, Wahab S, Vora LK, Singh Thakur RR, Kesharwani P. The cubosome-based nanoplatforms in cancer therapy: Seeking new paradigms for cancer theranostics. ENVIRONMENTAL RESEARCH 2023; 237:116894. [PMID: 37586450 DOI: 10.1016/j.envres.2023.116894] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/10/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023]
Abstract
Lyotropic liquid crystals are self-assembled, non-lamellar, and mesophase nanostructured materials that have garnered significant attention as drug carriers. Cubosomes, a subtype of lyotropic liquid crystalline nanoparticles, possess three-dimensional structures that display bicontinuous cubic liquid-crystalline patterns. These patterns are formed through the self-organization of unsaturated monoglycerides (amphphilic lipids such as glyceryl monooleate or phytantriol), followed by stabilization using steric polymers (poloxamers). Owing to their bicontinuous structure and steric polymer-based stabilization, cubosomes have been demonstrated to possess greater entrapment efficiency for hydrophobic drugs compared to liposomes, while also exhibiting high stability. In the past decade, there has been significant interest in cubosomes due to their ability to deliver therapeutic and contrast agents for cancer treatment and imaging with minimal side effects, establishing them as a safe and effective approach. Concerning these advantages, the present review elaborates on the general aspects, composition, and preparation techniques of cubosomes, followed by explanations of their mechanisms of drug loading and release patterns. Furthermore, the review provides meticulous discussions on the use of cubosomes in the treatment and imaging of various types of cancer, culminating in the enumeration of patents related to cubosome-based drug delivery systems.
Collapse
Affiliation(s)
- B H Jaswanth Gowda
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, Karnataka, India
| | - Mohammed Gulzar Ahmed
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, Karnataka, India
| | - Saad Ali Alshehri
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, United Kingdom
| | - Raghu Raj Singh Thakur
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast, BT9 7BL, United Kingdom
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India; Center for Global health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India.
| |
Collapse
|
21
|
Beniwal SS, Lamo P, Kaushik A, Lorenzo-Villegas DL, Liu Y, MohanaSundaram A. Current Status and Emerging Trends in Colorectal Cancer Screening and Diagnostics. BIOSENSORS 2023; 13:926. [PMID: 37887119 PMCID: PMC10605407 DOI: 10.3390/bios13100926] [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: 08/29/2023] [Revised: 09/27/2023] [Accepted: 10/07/2023] [Indexed: 10/28/2023]
Abstract
Colorectal cancer (CRC) is a prevalent and potentially fatal disease categorized based on its high incidences and mortality rates, which raised the need for effective diagnostic strategies for the early detection and management of CRC. While there are several conventional cancer diagnostics available, they have certain limitations that hinder their effectiveness. Significant research efforts are currently being dedicated to elucidating novel methodologies that aim at comprehending the intricate molecular mechanism that underlies CRC. Recently, microfluidic diagnostics have emerged as a pivotal solution, offering non-invasive approaches to real-time monitoring of disease progression and treatment response. Microfluidic devices enable the integration of multiple sample preparation steps into a single platform, which speeds up processing and improves sensitivity. Such advancements in diagnostic technologies hold immense promise for revolutionizing the field of CRC diagnosis and enabling efficient detection and monitoring strategies. This article elucidates several of the latest developments in microfluidic technology for CRC diagnostics. In addition to the advancements in microfluidic technology for CRC diagnostics, the integration of artificial intelligence (AI) holds great promise for further enhancing diagnostic capabilities. Advancements in microfluidic systems and AI-driven approaches can revolutionize colorectal cancer diagnostics, offering accurate, efficient, and personalized strategies to improve patient outcomes and transform cancer management.
Collapse
Affiliation(s)
| | - Paula Lamo
- Escuela Superior de Ingeniería y Tecnología, Universidad Internacional de La Rioja, 26006 Logroño, Spain
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Department of Environmental Engineering, Florida Polytechnic University, Lakeland, FL 33805, USA
| | | | - Yuguang Liu
- Departments of Physiology and Biomedical Engineering, Immunology and Surgery, Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
22
|
Yue NN, Xu HM, Xu J, Zhu MZ, Zhang Y, Tian CM, Nie YQ, Yao J, Liang YJ, Li DF, Wang LS. Application of Nanoparticles in the Diagnosis of Gastrointestinal Diseases: A Complete Future Perspective. Int J Nanomedicine 2023; 18:4143-4170. [PMID: 37525691 PMCID: PMC10387254 DOI: 10.2147/ijn.s413141] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/02/2023] [Indexed: 08/02/2023] Open
Abstract
The diagnosis of gastrointestinal (GI) diseases currently relies primarily on invasive procedures like digestive endoscopy. However, these procedures can cause discomfort, respiratory issues, and bacterial infections in patients, both during and after the examination. In recent years, nanomedicine has emerged as a promising field, providing significant advancements in diagnostic techniques. Nanoprobes, in particular, offer distinct advantages, such as high specificity and sensitivity in detecting GI diseases. Integration of nanoprobes with advanced imaging techniques, such as nuclear magnetic resonance, optical fluorescence imaging, tomography, and optical correlation tomography, has significantly enhanced the detection capabilities for GI tumors and inflammatory bowel disease (IBD). This synergy enables early diagnosis and precise staging of GI disorders. Among the nanoparticles investigated for clinical applications, superparamagnetic iron oxide, quantum dots, single carbon nanotubes, and nanocages have emerged as extensively studied and utilized agents. This review aimed to provide insights into the potential applications of nanoparticles in modern imaging techniques, with a specific focus on their role in facilitating early and specific diagnosis of a range of GI disorders, including IBD and colorectal cancer (CRC). Additionally, we discussed the challenges associated with the implementation of nanotechnology-based GI diagnostics and explored future prospects for translation in this promising field.
Collapse
Affiliation(s)
- Ning-ning Yue
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| | - Hao-ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, People’s Republic of China
| | - Jing Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, People’s Republic of China
| | - Min-zheng Zhu
- Department of Gastroenterology and Hepatology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, Guangdong, People’s Republic of China
| | - Cheng-Mei Tian
- Department of Emergency, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| | - Yu-qiang Nie
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, People’s Republic of China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| | - Yu-jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen, Guangdong, People’s Republic of China
| | - De-feng Li
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| | - Li-sheng Wang
- Department of Gastroenterology, Shenzhen People’s Hospital (the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
23
|
Vashishat A, Singh A, Kurmi BD, Gupta GD, Singh D. A short appraisal of polylactic-co-glycolic acid based polymer nanotechnology for colon cancer: recent advances and literature evidences. Ther Deliv 2023; 14:459-472. [PMID: 37559461 DOI: 10.4155/tde-2023-0027] [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: 08/11/2023] Open
Abstract
The currently available formulations provided non-targeted treatment of colon cancer, the deadliest cancer variant. Due to biopharmaceutical hindrances, the majority of the drugs are unable to reach the target site. Polylactic-co-glycolic acid (PLGA) is one of the versatile polymers in cancer treatment, diagnostics and theranostics. The unique mechanism of surface modifications in PLGA properties in colon cancer has been a keen interest to be used in different nanoparticles for improving biopharmaceutical attributes. The ongoing use of these smart nano-carriers has allowed targeted delivery of several active components on a wide scale. The main goal of this review is to compile information on PLGA-based nanocarriers which possess several desirable properties for drug delivery applications, including biocompatibility, biodegradability and tunable drug-release kinetics.
Collapse
Affiliation(s)
- Abhinav Vashishat
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, Punjab, 142001, India
| | - Amrinder Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140417, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, Punjab, 142001, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, Punjab, 142001, India
| | - Dilpreet Singh
- University Institute of Pharmaceutical Sciences, Chandigarh University, Gharuan, Mohali, 140413, India
| |
Collapse
|
24
|
Neto Í, Rocha J, Gaspar MM, Reis CP. Experimental Murine Models for Colorectal Cancer Research. Cancers (Basel) 2023; 15:2570. [PMID: 37174036 PMCID: PMC10177088 DOI: 10.3390/cancers15092570] [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: 03/29/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Colorectal cancer (CRC) is the third most prevalent malignancy worldwide and in both sexes. Numerous animal models for CRC have been established to study its biology, namely carcinogen-induced models (CIMs) and genetically engineered mouse models (GEMMs). CIMs are valuable for assessing colitis-related carcinogenesis and studying chemoprevention. On the other hand, CRC GEMMs have proven to be useful for evaluating the tumor microenvironment and systemic immune responses, which have contributed to the discovery of novel therapeutic approaches. Although metastatic disease can be induced by orthotopic injection of CRC cell lines, the resulting models are not representative of the full genetic diversity of the disease due to the limited number of cell lines suitable for this purpose. On the other hand, patient-derived xenografts (PDX) are the most reliable for preclinical drug development due to their ability to retain pathological and molecular characteristics. In this review, the authors discuss the various murine CRC models with a focus on their clinical relevance, benefits, and drawbacks. From all models discussed, murine CRC models will continue to be an important tool in advancing our understanding and treatment of this disease, but additional research is required to find a model that can correctly reflect the pathophysiology of CRC.
Collapse
Affiliation(s)
- Íris Neto
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (Í.N.); (J.R.)
| | - João Rocha
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (Í.N.); (J.R.)
| | - Maria Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (Í.N.); (J.R.)
| | - Catarina P. Reis
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (Í.N.); (J.R.)
- Instituto de Biofísica e Engenharia Biomédica (IBEB), Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| |
Collapse
|
25
|
Kim S, Lee W, Park H, Kim K. Tumor Microenvironment-Responsive 6-Mercaptopurine-Releasing Injectable Hydrogel for Colon Cancer Treatment. Gels 2023; 9:gels9040319. [PMID: 37102931 PMCID: PMC10138092 DOI: 10.3390/gels9040319] [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: 03/15/2023] [Revised: 04/02/2023] [Accepted: 04/08/2023] [Indexed: 04/28/2023] Open
Abstract
Colon cancer is a significant health concern. The development of effective drug delivery systems is critical for improving treatment outcomes. In this study, we developed a drug delivery system for colon cancer treatment by embedding 6-mercaptopurine (6-MP), an anticancer drug, in a thiolated gelatin/polyethylene glycol diacrylate hydrogel (6MP-GPGel). The 6MP-GPGel continuously released 6-MP, the anticancer drug. The release rate of 6-MP was further accelerated in an acidic or glutathione environment that mimicked a tumor microenvironment. In addition, when pure 6-MP was used for treatment, cancer cells proliferated again from day 5, whereas a continuous supply of 6-MP from the 6MP-GPGel continuously suppressed the survival rate of cancer cells. In conclusion, our study demonstrates that embedding 6-MP in a hydrogel formulation can improve the efficacy of colon cancer treatment and may serve as a promising minimally invasive and localized drug delivery system for future development.
Collapse
Affiliation(s)
- Sungjun Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, 30, Pildong-ro 1-gil, Jung-gu, Seoul 22012, Republic of Korea
| | - Wonjeong Lee
- Department of Chemical & Biochemical Engineering, Dongguk University, 30, Pildong-ro 1-gil, Jung-gu, Seoul 22012, Republic of Korea
| | - Heewon Park
- Department of Chemical & Biochemical Engineering, Dongguk University, 30, Pildong-ro 1-gil, Jung-gu, Seoul 22012, Republic of Korea
| | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, 30, Pildong-ro 1-gil, Jung-gu, Seoul 22012, Republic of Korea
| |
Collapse
|
26
|
Kumar S, Shukla MK, Sharma AK, Jayaprakash GK, Tonk RK, Chellappan DK, Singh SK, Dua K, Ahmed F, Bhattacharyya S, Kumar D. Metal-based nanomaterials and nanocomposites as promising frontier in cancer chemotherapy. MedComm (Beijing) 2023; 4:e253. [PMID: 37025253 PMCID: PMC10072971 DOI: 10.1002/mco2.253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 03/05/2023] [Accepted: 03/09/2023] [Indexed: 04/07/2023] Open
Abstract
Cancer is a disease associated with complex pathology and one of the most prevalent and leading reasons for mortality in the world. Current chemotherapy has challenges with cytotoxicity, selectivity, multidrug resistance, and the formation of stemlike cells. Nanomaterials (NMs) have unique properties that make them useful for various diagnostic and therapeutic purposes in cancer research. NMs can be engineered to target cancer cells for early detection and can deliver drugs directly to cancer cells, reducing side effects and improving treatment efficacy. Several of NMs can also be used for photothermal therapy to destroy cancer cells or enhance immune response to cancer by delivering immune-stimulating molecules to immune cells or modulating the tumor microenvironment. NMs are being modified to overcome issues, such as toxicity, lack of selectivity, increase drug capacity, and bioavailability, for a wide spectrum of cancer therapies. To improve targeted drug delivery using nano-carriers, noteworthy research is required. Several metal-based NMs have been studied with the expectation of finding a cure for cancer treatment. In this review, the current development and the potential of plant and metal-based NMs with their effects on size and shape have been discussed along with their more effective usage in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Sunil Kumar
- Department of Pharmaceutical ChemistrySchool of Pharmaceutical SciencesShoolini UniversitySolanHimachal PradeshIndia
| | - Monu Kumar Shukla
- Department of Pharmaceutical ChemistrySchool of Pharmaceutical SciencesShoolini UniversitySolanHimachal PradeshIndia
| | | | | | - Rajiv K. Tonk
- School of Pharmaceutical SciencesDelhi Pharmaceutical Sciences and Research UniversityNew DelhiDelhiIndia
| | | | - Sachin Kumar Singh
- School of Pharmaceutical SciencesLovely Professional UniversityPhagwaraPunjabIndia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of HealthUniversity of Technology SydneyUltimoNew South WalesAustralia
- Discipline of Pharmacy, Graduate School of Health, University of Technology SydneySydneyAustralia
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneySydneyAustralia
| | - Faheem Ahmed
- Department of PhysicsCollege of ScienceKing Faisal UniversityAl‐HofufAl‐AhsaSaudi Arabia
| | | | - Deepak Kumar
- Department of Pharmaceutical ChemistrySchool of Pharmaceutical SciencesShoolini UniversitySolanHimachal PradeshIndia
| |
Collapse
|
27
|
Liang Z, Gao J, Yin ZZ, Li J, Cai W, Kong Y. A sequential delivery system based on MoS 2 nanoflower doped chitosan/oxidized dextran hydrogels for colon cancer treatment. Int J Biol Macromol 2023; 233:123616. [PMID: 36773878 DOI: 10.1016/j.ijbiomac.2023.123616] [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: 12/17/2022] [Revised: 01/24/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023]
Abstract
A sequential delivery system based on MoS2 nanoflower (MoS2 NF) doped chitosan (CS)/oxidized dextran (OD) hydrogels is developed for the treatment of colon cancer. 5-Fluorouracil (5-FU) is combined with polyethylenimine (PEI) decorated MoS2 NF via electrostatic attraction and hydrogen bonding, and the obtained 5-FU/PEI/MoS2 is encapsulated by 1-tetradecanol (TD), a commonly used phase transition material. The resultant TD/5-FU/PEI/MoS2 (TFPM) is then co-encapsulated with methotrexate (MTX) in the CS/OD hydrogels generated via Schiff base reaction and electrostatic attraction. Because the electrostatic attraction between CS and OD is pH-sensitive, MTX and TD/5-FU/PEI/MoS2 can be easily released from the hydrogels at pH 7.4. MoS2 is an outstanding photothermal agent, and the generated hyperthermia under near infrared (NIR) irradiation can lead to the melting of TD and the consequent release of 5-FU encapsulated. More importantly, the generated hyperthermia under NIR irradiation can realize the chemo-photothermal synergistic tumor therapy. Finally, the practicability of the developed sequential delivery system is demonstrated by cytotoxicity test.
Collapse
Affiliation(s)
- Zhengyin Liang
- Jiangsu Key Laboratory of Advanced Catalytic Materials and Technology, School of Petrochemical Engineering, Changzhou University, Changzhou 213164, China
| | - Jun Gao
- Department of Orthopedics, Changzhou Municipal Hospital of Traditional Chinese Medicine, Changzhou 213003, China.
| | - Zheng-Zhi Yin
- College of Biological, Chemical Sciences and Engineering, Jiaxing University, Jiaxing 314001, China
| | - Junyao Li
- Jiangsu Key Laboratory of Advanced Catalytic Materials and Technology, School of Petrochemical Engineering, Changzhou University, Changzhou 213164, China
| | - Wenrong Cai
- Jiangsu Key Laboratory of Advanced Catalytic Materials and Technology, School of Petrochemical Engineering, Changzhou University, Changzhou 213164, China
| | - Yong Kong
- Jiangsu Key Laboratory of Advanced Catalytic Materials and Technology, School of Petrochemical Engineering, Changzhou University, Changzhou 213164, China.
| |
Collapse
|
28
|
Simón M, Jørgensen JT, Norregaard K, Henriksen JR, Clergeaud G, Andresen TL, Hansen AE, Kjaer A. Neoadjuvant Gold Nanoshell-Based Photothermal Therapy Combined with Liposomal Doxorubicin in a Mouse Model of Colorectal Cancer. Int J Nanomedicine 2023; 18:829-841. [PMID: 36824412 PMCID: PMC9942687 DOI: 10.2147/ijn.s389260] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/23/2022] [Indexed: 02/18/2023] Open
Abstract
Introduction Traditional cancer treatments, such as chemotherapy, are often incapable of achieving complete responses as standalone therapies. Hence, current treatment strategies typically rely on a combination of several approaches. Nanoparticle-based photothermal therapy (PTT) is a technique used to kill cancer cells through localized, severe hyperthermia that has shown promise as an add-on treatment to multiple cancer therapies. Here, we evaluated whether the combination of gold nanoshell (NS)-based PTT and liposomal doxorubicin could improve outcome in a mouse model of colorectal cancer. Methods First, NS-based PTT was performed on tumor-bearing mice. Radiolabeled liposomes were then injected at different timepoints to follow their accumulation in the tumor and determine the ideal injection time after PTT. In addition, fluorescent liposomes were used to observe the liposomal distribution in the tumor after PTT. Finally, we combined PTT and doxorubicin-loaded liposomes and studied the effect of the treatment strategy on the mice by following tumor growth and survival. Results PTT significantly improved liposomal accumulation in the tumor, but only when the liposomes were injected immediately after the therapy. The liposomes accumulated mostly in regions adjacent to the ablated areas. When PTT was combined with liposomal doxorubicin, the mice experienced a slowdown in tumor growth and an improvement in survival. Conclusion According to our preclinical study, NS-based PTT seems promising as an add-on treatment for liposomal chemotherapy and potentially other systemic therapies, and could be relevant for future application in a clinical setting.
Collapse
Affiliation(s)
- Marina Simón
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital – Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Tranekjær Jørgensen
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital – Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kamilla Norregaard
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital – Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonas Rosager Henriksen
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Gael Clergeaud
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Thomas L Andresen
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Anders Elias Hansen
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital – Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark,Correspondence: Andreas Kjaer, Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital – Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark, Email
| |
Collapse
|
29
|
Dezhakam E, Khalilzadeh B, Mahdipour M, Isildak I, Yousefi H, Ahmadi M, Naseri A, Rahbarghazi R. Electrochemical biosensors in exosome analysis; a short journey to the present and future trends in early-stage evaluation of cancers. Biosens Bioelectron 2023; 222:114980. [PMID: 36521207 DOI: 10.1016/j.bios.2022.114980] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/15/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
The tumor microenvironment consists of a multiplicity of cells such as cancer cells, fibroblasts, endothelial cells, and immune cells within the specific parenchyma. It has been indicated that cancer cells can educate other cells within the tumor niche in a paracrine manner by the release of nano-sized extracellular vesicles namely exosomes (Exo), resulting in accelerated tumor mass growth. It is suggested that exosomal cargo with remarkable information can reflect any changes in metabolic and proteomic profiles in parent tumor cells. Therefore, exosomes can be touted as prognostic, diagnostic, and therapeutic elements with specific biomarkers in patients with different tumor types. Despite the advantages, conventional exosome separation and purification protocols are time-consuming and laborious with low abnormal morphology and purity rate. During the last decades, biosensor-based modalities, as emerging instruments, have been used to detect and analyze Exo in biofluids. Due to suitable specificity, sensitivity, and real-time readout, biosensors became promising approaches for the analysis of Exo in in vitro and in vivo settings. The inherent advantages and superiority of electrochemical biosensors in the determination of tumor grade based on exosomal cargo and profile were also debated. Present and future challenges were also discussed related to the application of electrochemical biosensors in the clinical setting. In this review, the early detection of several cancer types associated with ovaries, breast, brain, colon, lungs, T and B lymphocytes, liver and rare types of cancers were debated in association with released exosomes.
Collapse
Affiliation(s)
- Ehsan Dezhakam
- Department of Analytical Chemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran
| | - Balal Khalilzadeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ibrahim Isildak
- Department of Bioengineering, Faculty of Chemistry-Metallurgy, Yildiz Technical University, 34220, Istanbul, Turkey
| | - Hadi Yousefi
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| | - Mahdi Ahmadi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abdolhossein Naseri
- Department of Analytical Chemistry, Faculty of Chemistry, University of Tabriz, Tabriz, Iran.
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
30
|
Yang B, Wang L, Tian Z. Silencing of RhoC induces macrophage M1 polarization to inhibit migration and invasion in colon cancer via regulating the PTEN/FOXO1 pathway. Int J Exp Pathol 2023; 104:33-42. [PMID: 36576072 PMCID: PMC9845608 DOI: 10.1111/iep.12460] [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: 07/11/2022] [Revised: 09/01/2022] [Accepted: 10/18/2022] [Indexed: 12/29/2022] Open
Abstract
Ras homologue family member C (RhoC) is an oncogene in diverse types of human cancers, whereas its regulatory mechanisms involving macrophage polarization is rarely investigated. This study is designed to explore the regulatory role of RhoC in colon cancer and the underlying molecular mechanisms involving macrophage polarization. We detected RhoC expression by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot, and analysed the biological function of RhoC knockdown in CC cells by the MTT, wound healing and transwell assay. Macrophage polarization-associated markers, genes associated with migration, phosphatase and tensin homologue (PTEN) and forkhead box O (FOXO) were determined by qRT-PCR and western blot. The xenograft tumour mouse model was used to assess the role of RhoC in vivo. RhoC is highly expressed in CC cells. The cell viability, invasion and migration abilities of CC cells were reduced by knockdown of RhoC. RhoC knockdown promoted M1 polarization, inhibited M2 polarization and decreased levels of genes associated with migration (matrix metalloproteinase-2 and matrix metalloproteinase-9). Silencing of RhoC inhibited tumour growth and expression of genes associated with migration in the xenografted model. In addition, silencing of RhoC promoted PTEN/FOXO1 expression, and PTEN inhibitor (SF1670) reversed the inhibitory effects of RhoC silencing. We demonstrated that silencing of RhoC reduced CC cells invasion and migration, and tumour growth by suppressing M2 macrophage polarization via regulating the PTEN/FOXO1 pathway.
Collapse
Affiliation(s)
- Bin Yang
- Department of GastroenterologyHarrison International Peace HospitalHengshuiChina
| | - Lihua Wang
- Department of GastroenterologyHarrison International Peace HospitalHengshuiChina
| | - Zhiying Tian
- Department of GastroenterologyHarrison International Peace HospitalHengshuiChina
| |
Collapse
|
31
|
Hazarika B, Singh VP. Macrocyclic supramolecular biomaterials in anti-cancer therapeutics. CHINESE CHEM LETT 2023. [DOI: 10.1016/j.cclet.2023.108220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
|
32
|
Alyami NM, Almeer R, Alyami HM. Role of green synthesized platinum nanoparticles in cytotoxicity, oxidative stress, and apoptosis of human colon cancer cells (HCT-116). Heliyon 2022; 8:e11917. [PMID: 36506358 PMCID: PMC9732314 DOI: 10.1016/j.heliyon.2022.e11917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 08/12/2022] [Accepted: 11/18/2022] [Indexed: 11/27/2022] Open
Abstract
Progresses in the medicinal application of nanocompounds were accepted for the treatment of cancer. Nanoparticles-based therapy is of benefit for effective biodistribution and specific targeting. The current study investigated the anticancer effect of green synthesized platinum nanoparticles (PtNPs) against colon cancer cells (HCT-116). Flow cytometry and ELISA techniques were employed for detecting apoptotic and oxidative stress markers. Furthermore, PtNPs-lycopene (PtNPs-LP) on cell migration and invasion of HCT-116 cells was also examined. The PtNPs-LP was capable of diminishing cell proliferation and viability of HCT-116 cells in a dose-dependent mode. After treatment with PtNPs-LP, a significant increase in pro-apoptotic Bax and caspase-3 and a decrease in anti-apoptotic Bcl-2 was observed in treated cells that subsequently released cytochrome C into its cytoplasm, initiating cell death. Moreover, PtNPs-LP induced excessive generation of reactive oxygen species (ROS) and oxidative stress in cancer cells. In conclusion, PtNPs-LP exerts an antitumor effect against colon cancer cells via mediating important mechanisms such as cytotoxicity, apoptosis, and oxidative stress.
Collapse
Affiliation(s)
- Nouf M. Alyami
- Department of Zoology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia,Corresponding author.
| | - Rafa Almeer
- Department of Zoology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Hanadi M. Alyami
- Specialized Dentistry Department, King Fahad Medical City, Riyadh 11451, Saudi Arabia
| |
Collapse
|
33
|
Tiwari A, Gajbhiye V, Jain A, Verma A, Shaikh A, Salve R, Jain SK. Hyaluronic acid functionalized liposomes embedded in biodegradable beads for duo drugs delivery to oxaliplatin-resistant colon cancer. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
34
|
Chen S, Zhu F, Li B, Yang J, Yang T, Liu X, Zhang J, Zhao Y. Alkaline media‐sensitive nanocarrier based on carboxylated cyclodextrin for targeted delivery of anti‐colon drug. J Appl Polym Sci 2022. [DOI: 10.1002/app.53163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Shuai Chen
- College of Chemistry and Chemical Engineering Yunnan Normal University Kunming People's Republic of China
| | - Fang‐Dao Zhu
- College of Chemistry and Chemical Engineering Yunnan Normal University Kunming People's Republic of China
| | - Bi‐Lian Li
- College of Chemistry and Chemical Engineering Yunnan Normal University Kunming People's Republic of China
| | - Jian‐Mei Yang
- College of Chemistry and Chemical Engineering Yunnan Normal University Kunming People's Republic of China
| | - Tong Yang
- College of Chemistry and Chemical Engineering Yunnan Normal University Kunming People's Republic of China
| | - Xiao‐Qing Liu
- Shenzhen Kewode Technology Co., Ltd Shenzhen People's Republic of China
| | - Jin Zhang
- College of Chemistry and Chemical Engineering Yunnan Normal University Kunming People's Republic of China
| | - Yan Zhao
- College of Chemistry and Chemical Engineering Yunnan Normal University Kunming People's Republic of China
| |
Collapse
|
35
|
Zhang H, Yu Y, Li J, Gong P, Wang X, Li X, Cheng Y, Yu X, Zhang N, Zhang X. Changes of gut microbiota in colorectal cancer patients with Pentatrichomonas hominis infection. Front Cell Infect Microbiol 2022; 12:961974. [PMID: 36118043 PMCID: PMC9471007 DOI: 10.3389/fcimb.2022.961974] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 07/27/2022] [Indexed: 01/11/2023] Open
Abstract
Pentatrichomonas hominis is a parasitic trichomonads protozoa that parasitizes in the colon and cecum of humans and other animals. Our previous studies have demonstrated that infection with P. hominis is associated with the incidence of colon cancer (37.93%). However, the mechanism by which P. hominis infections increase the incidence of colon cancer remains unclear. Previous studies have suggested that certain parasites promote colon cancer by regulating gut microbiota. This study aimed to elucidate whether the association between P. hominis infections and the increased incidence of colon cancer is related to changes in gut microbiota. Therefore, the gut microbiota patients with colon cancer who were infected with P. hominis and uninfected patients with colon cancer were analyzed by 16S rRNA high-throughput sequencing. The results demonstrated that patients with colon cancer who were not infected with P. hominis showed increased gut bacterial diversity, a higher relative abundance of Alcaligenes sp., Leucobacter sp., Paraprevotella sp., Ruminococcaceae UCG-002, and a significant reduction in the abundance of Veillonella sp., compared to individuals without colon cancer. Additionally, the relative abundance of the Ruminococcaceae UCG-002 and the Eubacterium eligens groups was reduced, while the relative abundance of bacteria associated with colon cancer, including Flavonifractor sp., Lachnoclostridium sp., and the Ruminococcus gnavus group, increased significantly in patients with colon cancer who were infected with P. hominis, compared to those of uninfected patients with colon cancer. In conclusion, these results suggested that P. hominis infections may aggravate the development of colon cancer and the findings provide new insights for subsequent in-depth studies on the pathogenesis, diagnosis, and prevention of colon cancer.
Collapse
Affiliation(s)
- Hongbo Zhang
- Key Laboratory of Zoonosis Research by Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yanhui Yu
- Second Affiliated Hospital, Jilin University, Changchun, China
| | - Jianhua Li
- Key Laboratory of Zoonosis Research by Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Pengtao Gong
- Key Laboratory of Zoonosis Research by Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiaocen Wang
- Key Laboratory of Zoonosis Research by Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xin Li
- Key Laboratory of Zoonosis Research by Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yidan Cheng
- Key Laboratory of Zoonosis Research by Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiuyan Yu
- Clinical Laboratory, Jilin Cancer Hospital, Changchun, China
| | - Nan Zhang
- Key Laboratory of Zoonosis Research by Ministry of Education, Institute of Zoonosis, Jilin University, Changchun, China
- *Correspondence: Nan Zhang, ; Xichen Zhang,
| | - Xichen Zhang
- Key Laboratory of Zoonosis Research by Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- *Correspondence: Nan Zhang, ; Xichen Zhang,
| |
Collapse
|
36
|
Wang C, Xue F, Wang M, An L, Wu D, Tian Q. 2D Cu-Bipyridine MOF Nanosheet as an Agent for Colon Cancer Therapy: A Three-in-One Approach for Enhancing Chemodynamic Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:38604-38616. [PMID: 35979620 DOI: 10.1021/acsami.2c11999] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Chemodynamic therapy (CDT) is a highly tumor-specific and minimally invasive treatment that is widely used in cancer therapy. However, its therapeutic effect is limited by the poor efficiency of hydroxyl radical generation. In colon cancer in particular, the high expression of hydrogen sulfide (H2S), which has strong reducibility, results in the consumption of generated hydroxyl radicals, further weakening the efficacy of CDT. To overcome this problem, we developed a novel two-dimensional (2D) Cu-bipyridine metal-organic framework (MOF) nanosheet [Cu(bpy)2(OTf)2] for colon cancer CDT. The therapeutic effect of Cu(bpy)2(OTf)2 is enhanced based on three factors. First, the developed 2D Cu-MOF rapidly consumes H2S to inhibit the consumption of generated hydroxyl radicals. Second, the ultrasmall CuS generated after H2S depletion facilitates Fenton-like reactions. Third, the generated CuS exhibits good photothermal performance in the second near-infrared window, allowing for photothermal-enhanced CDT. The ability of Cu(bpy)2(OTf)2 to improve the CDT effect was demonstrated through both in vitro and in vivo experiments. This work demonstrates the applicability of 2D Cu-MOF in the CDT of colon cancer and provides a novel strategy for constructing CDT agents for colon cancer.
Collapse
Affiliation(s)
- Chengbin Wang
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
- Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors, Shanghai Normal University, Shanghai 200234, China
| | - Fengfeng Xue
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Mengxin Wang
- Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors, Shanghai Normal University, Shanghai 200234, China
| | - Lu An
- Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors, Shanghai Normal University, Shanghai 200234, China
| | - Dan Wu
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Qiwei Tian
- Shanghai Key Laboratory of Molecular Imaging, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| |
Collapse
|
37
|
Inhibition of Colon Cancer Recurrence via Exogenous TRAIL Delivery Using Gel-like Coacervate Microdroplets. Gels 2022; 8:gels8070427. [PMID: 35877512 PMCID: PMC9319433 DOI: 10.3390/gels8070427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/04/2022] [Accepted: 07/07/2022] [Indexed: 02/04/2023] Open
Abstract
Colon cancer (CC) belongs to the three major malignancies with a high recurrence rate. Therefore, a novel drug delivery system that can prevent CC recurrence while minimizing side effects is needed. Tumor-necrosis-factor-related apoptosis-inducing ligand (TRAIL) has recently been spotlighted as a protein drug that can induce apoptosis of cancer cells specifically. However, its short in vivo half-life is still a challenge to overcome. Hence, in this study, a gel-like mPEGylated coacervate (mPEG-Coa) delivery platform was developed through electrostatic interaction of mPEG-poly(ethylene arginylaspartate diglyceride) (mPEG-PEAD) and heparin for effective protection of cargo TRAIL, subsequently preserving its bioactivity. mPEG-Coa could protect cargo TRAIL against protease. Sustained release was observed for a long-term (14 days). In addition, recurrence of HCT-116 cells was suppressed when cells were treated with TRAIL-loaded mPEG-Coa for 7 days through long-term continuous supply of active TRAIL, whereas re-proliferation occurred in the bolus TRAIL-treated group. Taken together, these results suggest that our gel-like mPEG-Coa could be utilized as a functional delivery platform to suppress CC recurrence by exogenously supplying TRAIL for a long time with a single administration.
Collapse
|
38
|
Wang CY, Sun M, Fan Z, Du JZ. Intestine Enzyme-responsive Polysaccharide-based Hydrogel to Open Epithelial Tight Junctions for Oral Delivery of Imatinib against Colon Cancer. CHINESE JOURNAL OF POLYMER SCIENCE 2022. [DOI: 10.1007/s10118-022-2726-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
39
|
Zhang Y, Lin J, Zhuo Y, Zou Z, Li Y, Yang H, Xie W, Zeng J, Deng Y, Cai S, Ye J, Zou F, Zhong W. Untargeted metabolomics reveals alterations in the metabolic reprogramming of prostate cancer cells by double-stranded DNA-modified gold nanoparticles. BIOMATERIALS ADVANCES 2022; 135:212745. [PMID: 35929217 DOI: 10.1016/j.bioadv.2022.212745] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 02/14/2022] [Accepted: 02/28/2022] [Indexed: 06/15/2023]
Abstract
Metabolic reprogramming plays an important role in the development of prostate cancer (PCa). However, there are few reports on the effects of nanomaterials as vectors on cancer metabolic reprogramming. Herein, a type of nanoparticle with good biocompatibility was synthesized by modifying the double-stranded of DNA containing a sulfhydryl group on the surface of gold nanoparticles (AuNPs-dsDNA) through salt-aging conjugation methods. The resultant AuNPs-dsDNA complexes possessed low toxicity to PC3 and DU145 cells in vitro. There was also no obvious hepatorenal toxicity after intravenous injection of AuNPs-dsDNA complexes in vivo, which indicated that these nanoparticles had good biological compatibilities. We investigated their biological functions using prostate cancer cells. Seahorse assay showed that AuNPs-dsDNA complexes could increase glycolysis and glycolysis capacity both in PC3 and DU145 cells. We further detected the expression of glycolysis-related genes by qPCR assay, and found that PKM2, PDHA, and LDHA were significantly upregulated. Furthermore, untargeted metabolomics revealed that PC (18:2(9Z,12Z)/18:2(9Z,12Z)) and PC (18:0/18:2 (9Z,12Z)) levels were decreased and inosinic acid level was increased in PC3 cells. Whereas (3S,6E,10E)-1,6,10,14-Phytatetraen-3-ol, Plasmenyl-PE 36:5 and Cer (d18:2/18:2) were decreased, PE 21:3 and 1-pyrrolidinecarboxaldehyde were increased in DU145 cells after co-culturing with AuNPs-dsDNA. In summary, we found that AuNPs and AuNPs-dsDNA complexes possibly regulate the metabolic reprogramming of cancer cells mainly through the lipid metabolic pathways, which could compensate for the previously mentioned phenomenon of enhanced glycolysis and glycolysis capacity. This will provide an important theoretical basis for our future research on the characteristic targeted design of nanomaterials for cancer metabolism.
Collapse
Affiliation(s)
- Yixun Zhang
- Department of Urology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Jundong Lin
- Department of Urology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Yangjia Zhuo
- Department of Urology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Zhihao Zou
- Department of Urology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Yuejiao Li
- Department of Urology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China; Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Huikang Yang
- Department of Urology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China; Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Wenjie Xie
- Department of Urology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Jie Zeng
- Department of Urology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Yulin Deng
- Department of Urology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Shanghua Cai
- Department of Urology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Jianheng Ye
- Department of Urology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Fen Zou
- Department of Urology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China.
| | - Weide Zhong
- Department of Urology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China.
| |
Collapse
|
40
|
Chen L, Lin Y, Zhang Z, Yang R, Bai X, Liu Z, Luo Z, Zhou M, Zhong Z. A novel dual-prodrug carried by cyclodextrin inclusion complex for the targeting treatment of colon cancer. J Nanobiotechnology 2021; 19:329. [PMID: 34666761 PMCID: PMC8524854 DOI: 10.1186/s12951-021-01064-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND There is an obvious correlation between ulcerative colitis and colorectal cancer, and the risk of colorectal cancer in patients with ulcerative colitis is increasing. Therefore, the combination therapy of anti-inflammatory and anti-tumor drugs may show promising to inhibit colon cancer. 5-aminosalicylic acid (5-ASA) with anti-inflammatory function is effective for maintaining remission in patients with ulcerative colitis and may also reduce colorectal cancer risk. Histone deacetylase (HDAC) plays an essential role in the progression of colon cancer. Butyric acid (BA) is a kind of HDAC inhibitor and thus shows tumor suppression to colon cancer. However, the volatile and corrosive nature of BA presents challenges in practical application. In addition, its clinical application is limited due to its non-targeting ability and low bioavailability. We aimed to synthesize a novel dual-prodrug of 5-ASA and BA, referred as BBA, to synergistically inhibit colon cancer. Further, based on the fact that folate receptor (FR) is over-expressed in most solid tumors and it has been identified to be a cancer stem cell surface marker in colon cancer, we took folate as the targeting ligand and used carboxymethyl-β-cyclodextrin (CM-β-CD) to carry BBA and thus prepared a novel inclusion complex of BBA/FA-PEG-CM-β-CD. RESULTS It was found that BBA/FA-PEG-CM-β-CD showed significant inhibition in cell proliferation against colon cancer cells SW620. It showed a pro-longed in vivo circulation and mainly accumulated in tumor tissue. More importantly, BBA/FA-PEG-CM-β-CD gave great tumor suppression effect against nude mice bearing SW620 xenografts. CONCLUSIONS Therefore, BBA/FA-PEG-CM-β-CD may have clinical potential in colon cancer therapy.
Collapse
Affiliation(s)
- Lin Chen
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.,Nanchong Key Laboratory of Individualized Drug Therapy, Department of Pharmacy, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Yan Lin
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zijun Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Ruisheng Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiaosheng Bai
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhongbing Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhongling Luo
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Meiling Zhou
- Department of Pharmacy, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Zhirong Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
41
|
Huang H, Wang J, Zhang J, Cai J, Pi J, Xu JF. Inspirations of Cobalt Oxide Nanoparticle Based Anticancer Therapeutics. Pharmaceutics 2021; 13:pharmaceutics13101599. [PMID: 34683892 PMCID: PMC8538820 DOI: 10.3390/pharmaceutics13101599] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/05/2021] [Accepted: 09/24/2021] [Indexed: 01/18/2023] Open
Abstract
Cobalt is essential to the metabolism of all animals due to its key role in cobalamin, also known as vitamin B12, the primary biological reservoir of cobalt as an ultra-trace element. Current cancer treatment strategies, including chemotherapy and radiotherapy, have been seriously restricted by their side effects and low efficiency for a long time, which urges us to develop new technologies for more effective and much safer anticancer therapies. Novel nanotechnologies, based on different kinds of functional nanomaterials, have been proved to act as effective and promising strategies for anticancer treatment. Based on the important biological roles of cobalt, cobalt oxide nanoparticles (NPs) have been widely developed for their attractive biomedical applications, especially their potential for anticancer treatments due to their selective inhibition of cancer cells. Thus, more and more attention has been attracted to the preparation, characterization and anticancer investigation of cobalt oxide nanoparticles in recent years, which is expected to introduce novel anticancer treatment strategies. In this review, we summarize the synthesis methods of cobalt oxide nanoparticles to discuss the advantages and restrictions for their preparation. Moreover, we emphatically discuss the anticancer functions of cobalt oxide nanoparticles as well as their underlying mechanisms to promote the development of cobalt oxide nanoparticles for anticancer treatments, which might finally benefit the current anticancer therapeutics based on functional cobalt oxide nanoparticles.
Collapse
Affiliation(s)
- Huanshao Huang
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (H.H.); (J.W.); (J.Z.)
| | - Jiajun Wang
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (H.H.); (J.W.); (J.Z.)
| | - Junai Zhang
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (H.H.); (J.W.); (J.Z.)
| | - Jiye Cai
- Department of Chemistry, Jinan University, Guangzhou 510632, China;
| | - Jiang Pi
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (H.H.); (J.W.); (J.Z.)
- Correspondence: (J.P.); (J.-F.X.)
| | - Jun-Fa Xu
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (H.H.); (J.W.); (J.Z.)
- Correspondence: (J.P.); (J.-F.X.)
| |
Collapse
|