1
|
Tymińska A, Karska N, Skoniecka A, Zawrzykraj M, Banach-Kopeć A, Mania S, Zieliński J, Kondej K, Gurzawska-Comis K, Skowron PM, Tylingo R, Rodziewicz-Motowidło S, Pikuła M. A novel chitosan-peptide system for cartilage tissue engineering with adipose-derived stromal cells. Biomed Pharmacother 2024; 181:117683. [PMID: 39561590 DOI: 10.1016/j.biopha.2024.117683] [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: 08/23/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
The natural healing process of cartilage injuries often fails to fully restore the tissue's biological and mechanical functions. Cartilage grafts are costly and require surgical intervention, often associated with complications such as intraoperative infection and rejection by the recipient due to ischemia. Novel tissue engineering technologies aim to ideally fill the cartilage defect to prevent disease progression or regenerate damaged tissue. Despite many studies on designing biocompatible composites to stimulate chondrogenesis, only few focus on peptides and carriers that promote stem cell proliferation or differentiation to promote healing. Our research aimed to design a carbohydrate chitosan-based biomaterial to stimulate stem cells into the chondrogenesis pathway. Our strategy was to combine chitosan with a novel peptide (UG28) that sequence was based on the copin protein. The construct stimulated human adipose-derived stem cells (AD-SCs) cells to undergo chondrogenic differentiation. Chitosan 75/500 allows AD-SCs to grow and has no harmful effects on the cells. The combination of UG28 peptide with the chitosan composite offers promising properties for cell differentiation, indicating its potential for clinical applications in cartilage regeneration.
Collapse
Affiliation(s)
- Agata Tymińska
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Department of Anatomy, Faculty of Medicine, Medical University of Gdańsk, Gdańsk 80-211, Poland.
| | - Natalia Karska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Gdańsk 80-308, Poland
| | - Aneta Skoniecka
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Department of Anatomy, Faculty of Medicine, Medical University of Gdańsk, Gdańsk 80-211, Poland
| | - Małgorzata Zawrzykraj
- Division of Clinical Anatomy, Department of Anatomy, Medical University of Gdańsk, 80-211, Poland
| | - Adrianna Banach-Kopeć
- Department of Chemistry, Technology and Biotechnology of Food Gdańsk University of Technology, Gdańsk 80-233, Poland
| | - Szymon Mania
- Department of Chemistry, Technology and Biotechnology of Food Gdańsk University of Technology, Gdańsk 80-233, Poland
| | - Jacek Zieliński
- Department of Oncologic Surgery, Medical University of Gdańsk, Gdańsk 80-214, Poland
| | - Karolina Kondej
- Department of Plastic Surgery, Medical University of Gdańsk, Gdańsk 80-214, Poland
| | - Katarzyna Gurzawska-Comis
- Department of Dentistry and Oral Health, Aarhus University, Vennelyst Boulevard 9, Aarhus C DK-8000, Denmark
| | - Piotr M Skowron
- Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, 80-308, Poland
| | - Robert Tylingo
- Department of Chemistry, Technology and Biotechnology of Food Gdańsk University of Technology, Gdańsk 80-233, Poland
| | | | - Michał Pikuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Division of Embryology, Department of Anatomy, Faculty of Medicine, Medical University of Gdańsk, Gdańsk 80-211, Poland.
| |
Collapse
|
2
|
Said ZAS, Mohammed HS, Ibrahim S, Amer HH. Electrospun zinc oxide nanoscaffolds: a targeted and selective anticancer approach. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024:1-22. [PMID: 39508689 DOI: 10.1080/09205063.2024.2422698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024]
Abstract
This study aims to prepare, characterize, and evaluate zinc oxide nanoscaffolds (ZnO NSs) as a potential anticancer drug that selectively targets malignant cells while remaining non-toxic to normal cells. Electrospun NSs were fabricated and loaded with varying concentrations of ZnO nanoparticles (NPs). The uniform morphology of the fabricated samples was confirmed through Field Emission Scanning Electron Microscope (FESEM) imaging. Elemental composition was investigated using Energy Dispersive X-ray spectroscopy (EDX), Fourier Transform Infrared (FTIR), and X-ray diffraction (XRD) analyses. Biocompatibility and cytotoxicity were assessed using the (3-(4.5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay) (MTT) assay and flow cytometry. The water uptake and degradation properties of the electrospun NSs were also examined. Furthermore, a cumulative release profile was generated to assess the release behavior of ZnO NSs. The prepared ZnO NSs demonstrated negligible toxicity toward normal human dermal cells. Conversely, the four used concentrations of ZnO NSs displayed substantial cytotoxicity and induced apoptosis in various cancer cell lines. The observed effects were concentration-dependent. Notably, ZnO NSs 8% exhibited the most significant reduction in cell viability against the MCF7 cell line. The findings from this study indicate the potential of ZnO NSs as an effective anticancer agent, with the ZnO NSs 8% demonstrating the most pronounced impact. This research introduces a novel application of electrospun zinc oxide nanoscaffolds, demonstrating their capacity for selective anticancer activity, particularly against breast carcinoma, while preserving normal cell viability. The study presents a significant advancement in the use of nanomaterial for targeted cancer therapy.
Collapse
Affiliation(s)
- Zeinab A S Said
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Haitham S Mohammed
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Sara Ibrahim
- Basic Medical Science Department, Faculty of Dentistry, Al Ryada University for Science and Technology, Menoufia, Egypt
| | - Hanan H Amer
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
3
|
Ray E, Jadhav K, Kadian M, Sharma G, Sharma K, Jhilta A, Singh R, Kumar A, Verma RK. Inhalable chitosan-coated nano-assemblies potentiate niclosamide for targeted abrogation of non-small-cell lung cancer through dual modulation of autophagy and apoptosis. Int J Biol Macromol 2024; 279:135411. [PMID: 39245099 DOI: 10.1016/j.ijbiomac.2024.135411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/14/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
Lung carcinoma, particularly non-small-cell lung cancer (NSCLC), accounts for a significant portion of cancer-related deaths, with a fatality rate of approximately 19 %. Niclosamide (NIC), originally an anthelmintic drug, has attracted attention for its potential in disrupting cancer cells through various intracellular signaling pathways. However, its effectiveness is hampered by limited solubility, reducing its bioavailability. This study investigates the efficacy of NIC against lung cancer using inhalable hybrid nano-assemblies with chitosan-functionalized Poly (ε-caprolactone) (PCL) as a carrier for pulmonary delivery. The evaluation encompasses various aspects such as aerodynamic and physicochemical properties, drug release kinetics, cellular uptake, biocompatibility, cell migration, autophagic flux, and apoptotic cell death in A549 lung cancer cells. Increasing NIC dosage correlates with enhanced inhibition of cell proliferation, showing a dose-dependent profile (approximately 75 % inhibition efficiency at 20 μg/mL of NIC). Optimization of inhaled dosage and efficacy is conducted in a murine model of NNK-induced tumor-bearing lung cancer. Following inhalation, NIC-CS-PCL-NA demonstrates significant lung deposition, retention, and metabolic stability. Inhalable nano-assemblies promote autophagy flux and induce apoptotic cell death. Preclinical trials reveal substantial tumor regression with minimal adverse effects, underscoring the potential of inhalable NIC-based nano-formulation as a potent therapeutic approach for NSCLC, offering effective tumor targeting and killing capabilities.
Collapse
Affiliation(s)
- Eupa Ray
- Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, SAS Nagar, Punjab 140306, India; University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Krishna Jadhav
- Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, SAS Nagar, Punjab 140306, India
| | - Monika Kadian
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Garima Sharma
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Kritika Sharma
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Agrim Jhilta
- Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, SAS Nagar, Punjab 140306, India
| | - Raghuraj Singh
- Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, SAS Nagar, Punjab 140306, India
| | - Anil Kumar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India.
| | - Rahul Kumar Verma
- Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, SAS Nagar, Punjab 140306, India.
| |
Collapse
|
4
|
Deng Z, Liu H, Chen G, Deng H, Dong X, Wang L, Tao F, Dai F, Cheng Y. Coaxial nanofibrous aerogel featuring porous network-structured channels for ovarian cancer treatment by sustained release of chitosan oligosaccharide. Int J Biol Macromol 2024; 276:133824. [PMID: 39002906 DOI: 10.1016/j.ijbiomac.2024.133824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Ovarian cancer, the deadliest gynecological malignancy, primarily treated with chemotherapy. However, systemic chemotherapy often leads to severe toxic side effects and chemoresistance. Drug-loaded aerogels have emerged as a promising method for drug delivery, as they can improve drug solubility and bioavailability, control drug release, and reduce drug distribution in non-targeted tissues, thereby minimizing side effects. In this research, chitosan oligosaccharide (COS)-loaded nanofibers composite chitosan (CS) aerogels (COS-NFs/CS) with a porous network structure were created using nanofiber recombination and freeze-drying techniques. The core layer of the aerogel has a COS loading rate of 60 %, enabling the COS-NFs/CS aerogel to significantly inhibit the migration and proliferation of ovarian cancer cells (resulting in a decrease in the survival rate of ovarian cancer cells to 33.70 % after 48 h). The coaxial fiber's unique shell-core structure and the aerogel's porous network structure enable the COS-NFs/CS aerogels to release COS steadily and slowly over 30 days, effectively reducing the initial burst release of COS. Additionally, the COS-NFs/CS aerogels exhibit good biocompatibility, degradability (only retaining 18.52 % of their weight after 6 weeks of implantation), and promote angiogenesis, thus promoting wound healing post-oophorectomy. In conclusion, COS-NFs/CS aerogels show great potential for application in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Zhimin Deng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Hua Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Gantao Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Hongbing Deng
- Hubei Key Laboratory of Biomass Resource Chemistry and Environmental Biotechnology, School of Resource and Environmental Science, Wuhan University, Wuhan 430079, China
| | - Xiangyang Dong
- Hubei Key Laboratory of Biomass Resource Chemistry and Environmental Biotechnology, School of Resource and Environmental Science, Wuhan University, Wuhan 430079, China
| | - Linlin Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Fenghua Tao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Fangfang Dai
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China.
| |
Collapse
|
5
|
Tang W, Hou H, Wang H, Gao X, Zhao F, Di Y, Ji S, Ling P, Wang F, Sun F, Tan H. Methotrexate-Loaded Chitosan Oligosaccharide-ES2 for Targeted Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:44409-44427. [PMID: 39162197 DOI: 10.1021/acsami.4c06656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Cancer presents a significant health threat, necessitating the development of more precise, efficient, and less damaging treatment approaches. To address this challenge, we employed the 1-ethyl-(3-dimethyl aminopropyl) carbodiimide/N-hydroxy succinimide (EDC/NHS) catalytic system and utilized quaternized chitosan oligosaccharide (HTCOSC) as a drug carrier to construct a nanoparticle delivery system termed HTCOSC-cRGD-ES2-MTX (CREM). This system specifically targets integrin αvβ3 on tumor cell surfaces and enables simultaneous loading of the antiangiogenic agent ES2 (IVRRADRAAVP) and the chemotherapy drug methotrexate (MTX). Due to its amphiphilic properties, CREM self-assembles into nanoparticles in aqueous solution, exhibiting an average diameter of 179.47 nm. Comparative studies demonstrated that CREM, in contrast to free ES2 and MTX-free nanoparticles (CRE), significantly suppressed the proliferation of EAhy926 endothelial cells and B16 melanoma cells in vitro, resulting in inhibition rates of 71.18 and 82.25%, respectively. Furthermore, CREM exhibited a hemolysis rate below 2%, indicating excellent in vitro antiangiogenic and antitumor activity as well as favorable blood compatibility. Additionally, both CRE and CREM demonstrated favorable tumor targeting capabilities through the specific binding action of cyclic RGD (cRGD) to integrin αvβ3. Further in vivo investigations revealed that CREM induced apoptosis in tumor cells via the mitochondrial apoptotic pathway and reduced the expression of angiogenic factors such as vascular endothelial growth factor (VEGF), thereby inhibiting tumor angiogenesis. This potent antitumor effect was evident through a tumor suppression rate of 80.19%. Importantly, histopathological staining (HE staining) demonstrated the absence of significant toxic side effects of CREM on various organs compared to MTX. In conclusion, the CREM nano drug delivery system synergistically enhances the therapeutic efficacy of antiangiogenic drugs and chemotherapeutic agents, thus offering a novel targeted approach for cancer treatment.
Collapse
Affiliation(s)
- Wen Tang
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, Shandong, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao 266237, China
| | - Huiwen Hou
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, Shandong, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao 266237, China
| | - Hanlin Wang
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, Shandong, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao 266237, China
| | - Xinqing Gao
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, Shandong, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao 266237, China
| | - Feiyan Zhao
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, Shandong, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao 266237, China
| | - Yuhan Di
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, Shandong, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao 266237, China
| | - Shengli Ji
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, Shandong, China
- ReaLi Tide Biological Technology (Weihai) Co., Ltd, Weihai 264207, China
| | - Peixue Ling
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, Shandong, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao 266237, China
- School of Pharmaceutical sciences, Shandong University, Jinan 250012, China
| | - Fengshan Wang
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, Shandong, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao 266237, China
- School of Pharmaceutical sciences, Shandong University, Jinan 250012, China
| | - Feng Sun
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, Shandong, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao 266237, China
- School of Pharmaceutical sciences, Shandong University, Jinan 250012, China
| | - Haining Tan
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, Shandong, China
- NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based Medicine, Shandong University, Qingdao 266237, China
- Shandong Provincial Technology Innovation Center of Carbohydrate, Shandong University, Qingdao 266237, China
- School of Pharmaceutical sciences, Shandong University, Jinan 250012, China
| |
Collapse
|
6
|
Hashem AE, Elmasry IH, Lebda MA, El-Karim DRSG, Hagar M, Ebied SKM, Alotaibi BS, Rizk NI, Ghamry HI, Shukry M, Edres HA. Characterization and antioxidant activity of nano-formulated berberine and cyperus rotundus extracts with anti-inflammatory effects in mastitis-induced rats. Sci Rep 2024; 14:18462. [PMID: 39122736 PMCID: PMC11315693 DOI: 10.1038/s41598-024-66801-8] [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: 02/20/2024] [Accepted: 07/04/2024] [Indexed: 08/12/2024] Open
Abstract
Bovine mastitis caused by infectious pathogens, mainly Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli), constitutes a major destructive challenge for the dairy industry and public health. Berberine chloride (BER) and Cyperus rotundus possess a broad spectrum of anti-inflammatory, antioxidant, antibacterial, and antiproliferative activities; however, their bioavailability is low. This research aimed first to prepare an ethanolic extract of Cyperus rotundus rhizomes (CRE) followed by screening its phytochemical contents, then synthesis of BER and CRE loaded chitosan nanoparticles (NPs) (BER/CH-NPs and CRE/CH-NPs), afterward, the analysis of their loading efficiency in addition to the morphological and physicochemical characterization of the formulated NPs employing Scanning Electron Microscopy (SEM), Zeta Potential (ZP), Fourier Transform Infrared Spectroscopy (FTIR), Differential Scanning Calorimetry (DSC) and X-Ray Diffraction (XRD) assessments compared to their crude forms to evaluate the enhancement of bioavailability and stability. Isolation of bacterial strains from the milk of mastitic cows, used for induction of mammary gland (MG) inflammation in female albino rats, and a preliminary investigation of the prophylactic oral doses of the prepared NPs against S. aureus-induced mastitis in female rats. The minimal inhibitory concentration (MIC) of BER/CH-NPs and CRE/CH-NPs is 1 mg/kg b.w. BER/CH-NPs and CRE/CH-NPs alone or in combination show significant (P ≤ 0.05) DPPH radical scavenging activity (69.2, 88.5, and 98.2%, respectively) in vitro. Oral administration of BER/CH-NPs and CRE/CH-NPs to mastitis rats significantly (P ≤ 0.05) attenuated TNF-α (22.1, 28.6 pg/ml), IL-6 (33.4, 42.9 pg/ml), IL-18 (21.7, 34.7 pg/ml), IL-4 (432.9, 421.6 pg/ml), and MPO (87.1, 89.3 pg/ml) compared to mastitis group alongside the improvement of MG histopathological findings without any side effect on renal and hepatic functions. Despite promising results with BER and CRE nanoparticles, the study is limited by small-scale trials, a focus on acute administration, and partially explored nanoparticle-biological interactions, with no economic or scalability assessments. Future research should address these limitations by expanding trial scopes, exploring interactions further, extending study durations, and assessing economic and practical scalability. Field trials and regulatory compliance are also necessary to ensure practical application and safety in the dairy industry. In conclusion, the in vitro and in vivo results proved the antioxidant and anti-inflammatory efficacy of BER/CH-NPs and CRE/CH-NPs in low doses with minimal damage to the liver and kidney functions, supposing their promising uses in mastitis treatment.
Collapse
Affiliation(s)
- Aml E Hashem
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Ingi H Elmasry
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Mohamed A Lebda
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Dina R S Gad El-Karim
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Mohamed Hagar
- Department of Chemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
- Faculty of Advanced Basic Sciences, Alamein International University, Alamein City, Matrouh Governorate, Egypt
| | - Sawsan Kh M Ebied
- Bacteriology Unit, Animal Health Research Institute, Alexandria Province, Egypt
| | - Badriyah S Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia.
| | - Nermin I Rizk
- Medical Physiology Department, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Heba I Ghamry
- Nutrition and Food Science, Department of Biology, College of Science, King Khalid University, P.O. Box 960, Abha, 61421, Saudi Arabia
| | - Mustafa Shukry
- Department of Physiology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt.
| | - Hanan A Edres
- Department of Biochemistry, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
7
|
Iskandar A, Kim SK, Wong TW. “Drug-Free” chitosan nanoparticles as therapeutic for cancer treatment. POLYM REV 2024; 64:818-871. [DOI: 10.1080/15583724.2024.2323943] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 11/22/2023] [Accepted: 02/19/2024] [Indexed: 01/06/2025]
Affiliation(s)
- Athirah Iskandar
- Non-Destructive Biomedical and Pharmaceutical Research Centre, Smart Manufacturing Research Institute, Universiti Teknologi MARA Selangor, Puncak Alam, Malaysia
- Particle Design Research Group, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, Puncak Alam, Malaysia
| | - Se-Kwon Kim
- Department of Marine Sciences and Convergent Technology, Hanyang University, Ansan, Seoul, Republic of Korea
| | - Tin Wui Wong
- Non-Destructive Biomedical and Pharmaceutical Research Centre, Smart Manufacturing Research Institute, Universiti Teknologi MARA Selangor, Puncak Alam, Malaysia
- Particle Design Research Group, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, Puncak Alam, Malaysia
- Sino-Malaysia Molecular Oncology and Traditional Chinese Medicine Delivery Joint Research Centre, Medical College, Yangzhou University, Yangzhou, China
| |
Collapse
|
8
|
Wang X, Liu H, Fei Y, Song Z, Meng X, Yu J, Liu X, Li L, Qiu L, Qian Z, Zhou S, Wang X, Zhang H. Metabolic pathway-based subtyping reveals distinct microenvironmental states associated with diffuse large B-cell lymphoma outcomes. Hematol Oncol 2024; 42:e3279. [PMID: 38819002 DOI: 10.1002/hon.3279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/22/2024] [Accepted: 05/01/2024] [Indexed: 06/01/2024]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a biologically and clinically heterogeneous disease that requires personalized clinical treatment. Assigning patients to different risk categories and cytogenetic abnormality and genetic mutation groups has been widely applied for prognostic stratification of DLBCL. Increasing evidence has demonstrated that dysregulated metabolic processes contribute to the initiation and progression of DLBCL. Metabolic competition within the tumor microenvironment is also known to influence immune cell metabolism. However, metabolism- and immune-related stratification has not been established. Here, 1660 genes involved in 84 metabolic pathways were selected and tested to establish metabolic clusters (MECs) of DLBCL. MECs established based on independent lymphoma datasets distinguished different survival outcomes. The CIBERSORT algorithm and EcoTyper were applied to quantify the relative abundance of immune cell types and identify variation in cell states for 13 lineages comprising the tumor micro environment among different MECs, respectively. Functional characterization showed that MECs were an indicator of the immune microenvironment and correlated with distinctive mutational characteristics and oncogenic signaling pathways. The novel immune-related MECs exhibited promising clinical prognostic value and potential for informing DLBCL treatment decisions.
Collapse
Affiliation(s)
- Xiaohui Wang
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Hengqi Liu
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Yue Fei
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Zheng Song
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Xiangrui Meng
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Jingwei Yu
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Xia Liu
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Lanfang Li
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Lihua Qiu
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Zhengzi Qian
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Shiyong Zhou
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Xianhuo Wang
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Huilai Zhang
- National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine and Department of Lymphoma, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, The Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| |
Collapse
|
9
|
Schröder V, Gherghel D, Apetroaei MR, Gîjiu CL, Isopescu R, Dinculescu D, Apetroaei MM, Enache LE, Mihai CT, Rău I, Vochița G. α-Chitosan and β-Oligochitosan Mixtures-Based Formula for In Vitro Assessment of Melanocyte Cells Response. Int J Mol Sci 2024; 25:6768. [PMID: 38928474 PMCID: PMC11204147 DOI: 10.3390/ijms25126768] [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: 05/16/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Chitosan is a natural polymer with numerous biomedical applications. The cellular activity of chitosan has been studied in various types of cancer, including melanoma, and indicates that these molecules can open new perspectives on antiproliferative action and anticancer therapy. This study analyzes how different chitosan conformations, such as α-chitosan (CH) or β-oligochitosan (CO), with various degrees of deacetylation (DDA) and molar mass (MM), both in different concentrations and in CH-CO mixtures, influence the cellular processes of SK-MEL-28 melanocytes, to estimate the reactivity of these cells to the applied treatments. The in vitro evaluation was carried out, aiming at the cellular metabolism (MTT assay), cellular morphology, and chitinase-like glycoprotein YKL-40 expression. The in vitro effect of the CH-CO mixture application on melanocytes is obvious at low concentrations of α-chitosan/β-oligochitosan (1:2 ratio), with the cell's response supporting the hypothesis that β-oligo-chitosan amplifies the effect. This oligochitosan mixture, favored by the β conformation and its small size, penetrates faster into the cells, being more reactive when interacting with some cellular components. Morphological effects expressed by the loss of cell adhesion and the depletion of YKL-40 synthesis are significant responses of melanocytes. β-oligochitosan (1.5 kDa) induces an extension of cytophysiological effects and limits the cell viability compared to α-chitosan (400-900 kDa). Statistical analysis using multivariate techniques showed differences between the CH samples and CH-CO mixtures.
Collapse
Affiliation(s)
- Verginica Schröder
- Departament of Cellular and Molecular Biology, Faculty of Pharmacy, Ovidius University of Constanta, 6 Capt. Aviator Al. Șerbănescu Street, Campus C, 900470 Constanta, Romania;
| | - Daniela Gherghel
- Institute of Biological Research Iasi, Branch of NIRDBS—National Institute of Research and Development of Biological Sciences Bucharest, 47 Lascar Catargi, 700107 Iasi, Romania;
| | - Manuela Rossemary Apetroaei
- Department of Marine Electric and Electronic Engineering, Faculty of Marine Engineering, Mircea cel Batran Naval Academy, 1 Fulgerului Street, 900218 Constanta, Romania;
| | - Cristiana Luminița Gîjiu
- Faculty of Chemical Engineering and Biotechnologies, National University of Science and Technology POLITEHNICA Bucharest, 011061 Bucharest, Romania; (C.L.G.); (R.I.); (L.E.E.); (I.R.)
| | - Raluca Isopescu
- Faculty of Chemical Engineering and Biotechnologies, National University of Science and Technology POLITEHNICA Bucharest, 011061 Bucharest, Romania; (C.L.G.); (R.I.); (L.E.E.); (I.R.)
| | - Daniel Dinculescu
- Faculty of Chemical Engineering and Biotechnologies, National University of Science and Technology POLITEHNICA Bucharest, 011061 Bucharest, Romania; (C.L.G.); (R.I.); (L.E.E.); (I.R.)
| | - Miruna-Maria Apetroaei
- Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania;
| | - Laura Elena Enache
- Faculty of Chemical Engineering and Biotechnologies, National University of Science and Technology POLITEHNICA Bucharest, 011061 Bucharest, Romania; (C.L.G.); (R.I.); (L.E.E.); (I.R.)
| | | | - Ileana Rău
- Faculty of Chemical Engineering and Biotechnologies, National University of Science and Technology POLITEHNICA Bucharest, 011061 Bucharest, Romania; (C.L.G.); (R.I.); (L.E.E.); (I.R.)
| | - Gabriela Vochița
- Institute of Biological Research Iasi, Branch of NIRDBS—National Institute of Research and Development of Biological Sciences Bucharest, 47 Lascar Catargi, 700107 Iasi, Romania;
| |
Collapse
|
10
|
Amin H, Ibrahim IM, Hassanein EHM. Weaponizing chitosan and its derivatives in the battle against lung cancer. Int J Biol Macromol 2024; 272:132888. [PMID: 38844273 DOI: 10.1016/j.ijbiomac.2024.132888] [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: 12/27/2023] [Revised: 05/28/2024] [Accepted: 06/02/2024] [Indexed: 06/11/2024]
Abstract
Lung cancer (LC) is a crisis of catastrophic proportions. It is a global problem and urgently requires a solution. The classic chemo drugs are lagging behind as they lack selectivity, where their side effects are spilled all over the body, and these adverse effects would be terribly tragic for LC patients. Therefore, they could make a bad situation worse, inflict damage on normal cells, and inflict pain on patients. Since our confidence in classic drugs is eroding, chitosan can offer a major leap forward in LC therapy. It can provide the backbone and the vehicle that enable chemo drugs to penetrate the hard shell of LC. It could be functionalized in a variety of ways to deliver a deadly payload of toxins to kill the bad guys. It is implemented in formulation of polymeric NPs, lipidic NPs, nanocomposites, multiwalled carbon nanotubes, and phototherapeutic agents. This review is a pretty clear proof of chitosan's utility as a weapon in battling LC. Chitosan-based formulations could work effectively to kill LC cells. If a researcher is looking for a vehicle for medication for LC therapy, chitosan can be an appropriate choice.
Collapse
Affiliation(s)
- Haitham Amin
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt.
| | - Islam M Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt.
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt.
| |
Collapse
|
11
|
Elizalde-Cárdenas A, Ribas-Aparicio RM, Rodríguez-Martínez A, Leyva-Gómez G, Ríos-Castañeda C, González-Torres M. Advances in chitosan and chitosan derivatives for biomedical applications in tissue engineering: An updated review. Int J Biol Macromol 2024; 262:129999. [PMID: 38331080 DOI: 10.1016/j.ijbiomac.2024.129999] [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: 11/13/2023] [Revised: 01/19/2024] [Accepted: 02/04/2024] [Indexed: 02/10/2024]
Abstract
In recent years, chitosan (CS) has received much attention as a functional biopolymer for various applications, especially in the biomedical field. It is a natural polysaccharide created by the chemical deacetylation of chitin (CT) that is nontoxic, biocompatible, and biodegradable. This natural polymer is difficult to process; however, chemical modification of the CS backbone allows improved use of functional derivatives. CS and its derivatives are used to prepare hydrogels, membranes, scaffolds, fibers, foams, and sponges, primarily for regenerative medicine. Tissue engineering (TE), currently one of the fastest-growing fields in the life sciences, primarily aims to restore or replace lost or damaged organs and tissues using supports that, combined with cells and biomolecules, generate new tissue. In this sense, the growing interest in the application of biomaterials based on CS and some of its derivatives is justifiable. This review aims to summarize the most important recent advances in developing biomaterials based on CS and its derivatives and to study their synthesis, characterization, and applications in the biomedical field, especially in the TE area.
Collapse
Affiliation(s)
- Alejandro Elizalde-Cárdenas
- Conahcyt & Laboratorio de Biotecnología, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra", Ciudad de México 14389, Mexico; Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | - Rosa María Ribas-Aparicio
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | - Aurora Rodríguez-Martínez
- Conahcyt & Laboratorio de Biotecnología, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra", Ciudad de México 14389, Mexico; Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Camilo Ríos-Castañeda
- Dirección de investigación, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra", Ciudad de México 14389, Mexico
| | - Maykel González-Torres
- Conahcyt & Laboratorio de Biotecnología, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra", Ciudad de México 14389, Mexico.
| |
Collapse
|
12
|
Li T, Ashrafizadeh M, Shang Y, Nuri Ertas Y, Orive G. Chitosan-functionalized bioplatforms and hydrogels in breast cancer: immunotherapy, phototherapy and clinical perspectives. Drug Discov Today 2024; 29:103851. [PMID: 38092146 DOI: 10.1016/j.drudis.2023.103851] [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: 09/18/2023] [Revised: 11/12/2023] [Accepted: 12/01/2023] [Indexed: 12/19/2023]
Abstract
Breast cancer is the most common and malignant tumor among women. Chitosan (CS)-based nanoparticles have been introduced into breast cancer therapy as a way to increase the targeted delivery of drugs and genes to the tumor site. CS nanostructures suppress tumorigenesis by enhancing both the targeted delivery of cargo (drug and gene) and its accumulation in tumor cells. The tumor cells internalize CS-based nanoparticles through endocytosis. Moreover, chitosan nanocarriers can also induce phototherapy-mediated tumor ablation. Smart and multifunctional types of CS nanoparticles, including pH-, light- and redox-responsive nanoparticles, can be used to improve the potential for breast cancer removal. In addition, the acceleration of immunotherapy by CS nanoparticles has also been achieved, and there is potential to develop CS-nanoparticle hydrogels that can be used to suppress tumorigenesis.
Collapse
Affiliation(s)
- Tianfeng Li
- Reproductive Medicine Center, Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, 518055, China; Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China.
| | - Milad Ashrafizadeh
- Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China
| | - Yuru Shang
- Southern University of Science and Technology Hospital, Shenzhen 518055, China
| | - Yavuz Nuri Ertas
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, 38039, Turkey; Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey.
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; BTI-Biotechnology Institute, Vitoria, Spain; University Institute for Regenerative Medicine and Oral Implantology (UIRMI) (UPV/EHU-Fundación Eduardo Anitua), Vitoria-Gasteiz, Spain.
| |
Collapse
|
13
|
Gao Y, Luo L, Qu Y, Zhou Q. MFNG is an independent prognostic marker for osteosarcoma. Eur J Med Res 2023; 28:256. [PMID: 37496053 PMCID: PMC10369729 DOI: 10.1186/s40001-023-01139-x] [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: 07/07/2022] [Accepted: 05/13/2023] [Indexed: 07/28/2023] Open
Abstract
BACKGROUND Osteosarcoma (OS) has been the most common malignancy of the bone in children and adolescents, and the unsatisfactory prognosis of OS sufferers has long been a hard nut. Here, we delved into the markers with a prognostic value for predicting the prognosis of OS patients. METHODS The messenger RNA (mRNA) sequencing data and clinical data of OS were retrieved from a Gene Expression Omnibus (GEO) dataset (GSE39058). Next, prognosis-related genes (PRGs) were filtered with the aid of Kaplan-Meier (K-M) curves and Cox regression analysis (CRA). Later, Gene Ontology (GO) biological process analysis was used in verifying the function of different genes. CCK-8 and cell apoptosis assay were performed to evaluate the function of MFNG in U2OS cells. RESULTS Among the obtained genes, Manic Fringe (MFNG) had the closest relevance to prognosis and clinical traits, thus becoming the research object herein. In light of the expression level of MFNG, patients fell into high- and low-MFNG groups. Patients with elevated MFNG expression had a worse prognosis, according to the survival analysis. It was unveiled by the univariate and multivariate analyses that MFNG expression was an independent adverse prognostic factor for disease-free survival in OS patients (p = 0.006). Meanwhile, MFNG expression was linked to gender and tumor recurrence, and it was higher in patients with OS recurrence. Moreover, overexpression of MFNG promoted the cell proliferation and inhibited the cell apoptosis of U2OS cells. CONCLUSIONS The expression level of MFNG negatively correlated with OS progression, and as an independent adverse prognostic factor for disease-free survival in OS patients. Moreover, MFNG regulated the cell proliferation and apoptosis of OS cells.
Collapse
Affiliation(s)
- Yi Gao
- Department of Orthopaedics, Changzhou Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, 25 Heping Bei Lu, Tianning District, Changzhou, 213000, Jiangsu, China
| | - Lili Luo
- Department of Orthopaedics, Changzhou Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, 25 Heping Bei Lu, Tianning District, Changzhou, 213000, Jiangsu, China
| | - Yuxing Qu
- Department of Orthopaedics, Changzhou Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, 25 Heping Bei Lu, Tianning District, Changzhou, 213000, Jiangsu, China
| | - Qi Zhou
- Department of Orthopaedics, Changzhou Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, 25 Heping Bei Lu, Tianning District, Changzhou, 213000, Jiangsu, China.
| |
Collapse
|
14
|
Chen M, Lan H, Yao S, Jin K, Chen Y. Metabolic Interventions in Tumor Immunity: Focus on Dual Pathway Inhibitors. Cancers (Basel) 2023; 15:cancers15072043. [PMID: 37046703 PMCID: PMC10093048 DOI: 10.3390/cancers15072043] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023] Open
Abstract
The metabolism of tumors and immune cells in the tumor microenvironment (TME) can affect the fate of cancer and immune responses. Metabolic reprogramming can occur following the activation of metabolic-related signaling pathways, such as phosphoinositide 3-kinases (PI3Ks) and the mammalian target of rapamycin (mTOR). Moreover, various tumor-derived immunosuppressive metabolites following metabolic reprogramming also affect antitumor immune responses. Evidence shows that intervention in the metabolic pathways of tumors or immune cells can be an attractive and novel treatment option for cancer. For instance, administrating inhibitors of various signaling pathways, such as phosphoinositide 3-kinases (PI3Ks), can improve T cell-mediated antitumor immune responses. However, dual pathway inhibitors can significantly suppress tumor growth more than they inhibit each pathway separately. This review discusses the latest metabolic interventions by dual pathway inhibitors as well as the advantages and disadvantages of this therapeutic approach.
Collapse
Affiliation(s)
- Min Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Huanrong Lan
- Department of Surgical Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou 310002, China
| | - Shiya Yao
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Yun Chen
- Department of Colorectal Surgery, Xinchang People's Hospital, Affiliated Xinchang Hospital, Wenzhou Medical University, Xinchang 312500, China
| |
Collapse
|
15
|
Herdiana Y, Wathoni N, Gozali D, Shamsuddin S, Muchtaridi M. Chitosan-Based Nano-Smart Drug Delivery System in Breast Cancer Therapy. Pharmaceutics 2023; 15:pharmaceutics15030879. [PMID: 36986740 PMCID: PMC10051865 DOI: 10.3390/pharmaceutics15030879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 02/28/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
Despite recent advances, cancer remains the primary killer on a global scale. Numerous forms of research have been conducted to discover novel and efficient anticancer medications. The complexity of breast cancer is a major challenge which is coupled with patient-to-patient variations and heterogeneity between cells within the tumor. Revolutionary drug delivery is expected to provide a solution to that challenge. Chitosan nanoparticles (CSNPs) have prospects as a revolutionary delivery system capable of enhancing anticancer drug activity and reducing negative impacts on normal cells. The use of smart drug delivery systems (SDDs) as delivering materials to improve the bioactivity of NPs and to understand the intricacies of breast cancer has garnered significant interest. There are many reviews about CSNPs that present various points of view, but they have not yet described a series in cancer therapy from cell uptake to cell death. With this description, we will provide a more complete picture for designing preparations for SDDs. This review describes CSNPs as SDDSs, enhancing cancer therapy targeting and stimulus response using their anticancer mechanism. Multimodal chitosan SDDs as targeting and stimulus response medication delivery will improve therapeutic results.
Collapse
Affiliation(s)
- Yedi Herdiana
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
- Correspondence: (Y.H.); (M.M.)
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Dolih Gozali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Shaharum Shamsuddin
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
- Nanobiotech Research Initiative, Institute for Research in Molecular Medicine (INFORMM), USM, Penang 11800, Malaysia
- USM-RIKEN Interdisciplinary Collaboration on Advanced Sciences (URICAS), USM, Penang 11800, Malaysia
| | - Muchtaridi Muchtaridi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
- Correspondence: (Y.H.); (M.M.)
| |
Collapse
|
16
|
A Natural Glucan from Black Bean Inhibits Cancer Cell Proliferation via PI3K-Akt and MAPK Pathway. Molecules 2023; 28:molecules28041971. [PMID: 36838963 PMCID: PMC9961350 DOI: 10.3390/molecules28041971] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/12/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
A natural α-1,6-glucan named BBWPW was identified from black beans. Cell viability assay showed that BBWPW inhibited the proliferation of different cancer cells, especially HeLa cells. Flow cytometry analysis indicated that BBWPW suppressed the HeLa cell cycle in the G2/M phase. Consistently, RT-PCR experiments displayed that BBWPW significantly impacts the expression of four marker genes related to the G2/M phase, including p21, CDK1, Cyclin B1, and Survivin. To explore the molecular mechanism of BBWPW to induce cell cycle arrest, a transcriptome-based target inference approach was utilized to predict the potential upstream pathways of BBWPW and it was found that the PI3K-Akt and MAPK signal pathways had the potential to mediate the effects of BBWPW on the cell cycle. Further experimental tests confirmed that BBWPW increased the expression of BAD and AKT and decreased the expression of mTOR and MKK3. These results suggested that BBWPW could regulate the PI3K-Akt and MAPK pathways to induce cell cycle arrest and ultimately inhibit the proliferation of HeLa cells, providing the potential of the black bean glucan to be a natural anticancer drug.
Collapse
|
17
|
Zhang L, Li C, Marhaba Aziz, Zhu R, Jiapaer Z. ITF2357 induces cell cycle arrest and apoptosis of meningioma cells via the PI3K-Akt pathway. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 40:21. [PMID: 36445551 DOI: 10.1007/s12032-022-01883-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 11/02/2022] [Indexed: 12/02/2022]
Abstract
As a type of central nervous system tumor, meningioma usually compresses the nerve center due to its local expansion, further causing neurological deficits. However, there are limited therapeutic approaches for meningiomas. ITF2357, a potent class I and II histone deacetylase inhibitor (HDACi), has been shown to inhibit cell proliferation, promote apoptosis, and block the cell cycle in various sarcoma cells, including glioblastoma and peripheral T-cell lymphoma. Here, we investigated the potential role of ITF2357 on meningioma cancer cells (IOMM-Lee cells). First, we demonstrated that the half-maximal inhibitory concentration (IC50) of ITF2357 was 1.842 μM by MTT assay. In addition, ITF2357 effectively inhibited the proliferation and colonization ability of IOMM-Lee cells. Flow cytometry analysis showed that ITF2357 induced G0/G1 and G2/M phase cell cycle arrest and cell apoptosis. Mechanically, the RNA sequencing data revealed that ITF2357 could affect the PI3K-Akt signaling pathway and the cell cycle progression. Furthermore, the expression levels of Akt, PI3K, p-Akt, and p-PI3K were determined by western blotting. Collectively, our data revealed that ITF2357 induces G0 G1 and G2/M phase arrest and apoptosis by inhibiting hyperactivation of the PI3K-Akt pathway, ultimately inhibiting cell viability and proliferation of meningioma cells, which developed a new approach to the treatment of meningioma.
Collapse
Affiliation(s)
- Lingying Zhang
- Xinjiang Key Laboratory of Biological Resources and Gentic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830046, China
| | - Chengyu Li
- Xinjiang Key Laboratory of Biological Resources and Gentic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830046, China
| | - Marhaba Aziz
- Xinjiang Key Laboratory of Biological Resources and Gentic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830046, China
| | - Rongxin Zhu
- Xinjiang Key Laboratory of Biological Resources and Gentic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830046, China
| | - Zeyidan Jiapaer
- Xinjiang Key Laboratory of Biological Resources and Gentic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830046, China.
| |
Collapse
|
18
|
Chitosan-based therapeutic systems and their potentials in treatment of oral diseases. Int J Biol Macromol 2022; 222:3178-3194. [DOI: 10.1016/j.ijbiomac.2022.10.090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/09/2022] [Accepted: 10/10/2022] [Indexed: 11/05/2022]
|
19
|
Chen Q, Qi Y, Jiang Y, Quan W, Luo H, Wu K, Li S, Ouyang Q. Progress in Research of Chitosan Chemical Modification Technologies and Their Applications. Mar Drugs 2022; 20:md20080536. [PMID: 36005539 PMCID: PMC9410415 DOI: 10.3390/md20080536] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/14/2022] [Accepted: 08/16/2022] [Indexed: 02/07/2023] Open
Abstract
Chitosan, which is derived from chitin, is the only known natural alkaline cationic polymer. Chitosan is a biological material that can significantly improve the living standard of the country. It has excellent properties such as good biodegradability, biocompatibility, and cell affinity, and has excellent biological activities such as antibacterial, antioxidant, and hemostasis. In recent years, the demand has increased significantly in many fields and has huge application potential. Due to the poor water solubility of chitosan, its wide application is limited. However, chemical modification of the chitosan matrix structure can improve its solubility and biological activity, thereby expanding its application range. The review covers the period from 1996 to 2022 and was elaborated by searching Google Scholar, PubMed, Elsevier, ACS publications, MDPI, Web of Science, Springer, and other databases. The various chemical modification methods of chitosan and its main activities and application research progress were reviewed. In general, the modification of chitosan and the application of its derivatives have had great progress, such as various reactions, optimization of conditions, new synthetic routes, and synthesis of various novel multifunctional chitosan derivatives. The chemical properties of modified chitosan are usually better than those of unmodified chitosan, so chitosan derivatives have been widely used and have more promising prospects. This paper aims to explore the latest progress in chitosan chemical modification technologies and analyze the application of chitosan and its derivatives in various fields, including pharmaceuticals and textiles, thus providing a basis for further development and utilization of chitosan.
Collapse
Affiliation(s)
- Qizhou Chen
- The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China
| | - Yi Qi
- The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, Zhanjiang 524023, China
| | - Yuwei Jiang
- The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China
| | - Weiyan Quan
- The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, Zhanjiang 524023, China
| | - Hui Luo
- The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, Zhanjiang 524023, China
- Correspondence: (H.L.); (Q.O.); Tel.: +86-137-0273-9877 (H.L.); +86-180-2842-0107 (Q.O.)
| | - Kefeng Wu
- The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, Zhanjiang 524023, China
| | - Sidong Li
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, Zhanjiang 524023, China
| | - Qianqian Ouyang
- The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, Zhanjiang 524023, China
- Correspondence: (H.L.); (Q.O.); Tel.: +86-137-0273-9877 (H.L.); +86-180-2842-0107 (Q.O.)
| |
Collapse
|
20
|
Yuan J, Ye Z, Zeng Y, Pan Z, Feng Z, Bao Y, Li Y, Liu X, He Y, Feng Q. Bifunctional scaffolds for tumor therapy and bone regeneration: Synergistic effect and interplay between therapeutic agents and scaffold materials. Mater Today Bio 2022; 15:100318. [PMID: 35734197 PMCID: PMC9207581 DOI: 10.1016/j.mtbio.2022.100318] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 10/26/2022] Open
Abstract
Bone tumor patients often face the problems with cancer cell residues and bone defects after the operation. Therefore, researchers have developed many bifunctional scaffolds with both tumor treatment and bone repair functions. Therapeutic agents are usually combined with bioactive scaffolds to achieve the "bifunctional". However, the synergistic effect of bifunctional scaffolds on tumor therapy and bone repair, as well as the interplay between therapeutic agents and scaffold materials in bifunctional scaffolds, have not been emphasized and discussed. This review proposes a promising design scheme for bifunctional scaffolds: the synergistic effect and interplay between the therapeutic agents and scaffold materials. This review summarizes the latest research progress in bifunctional scaffolds for therapeutic applications and regeneration. In particular, it summarizes the role of tumor therapeutic agents in bone regeneration and the role of scaffold materials in tumor treatment. Finally, a perspective on the future development of bifunctional scaffolds for tumor therapy and bone regeneration is discussed.
Collapse
Affiliation(s)
- Jiongpeng Yuan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Zhaoyi Ye
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Yaoxun Zeng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Zhenxing Pan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - ZhenZhen Feng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Ying Bao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Yushan Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Xujie Liu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Yan He
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Qingling Feng
- School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
21
|
Chitooligosaccharides Improve the Efficacy of Checkpoint Inhibitors in a Mouse Model of Lung Cancer. Pharmaceutics 2022; 14:pharmaceutics14051046. [PMID: 35631632 PMCID: PMC9147765 DOI: 10.3390/pharmaceutics14051046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 12/10/2022] Open
Abstract
YKL-40 (also named chitinase 3 like-1 protein [CHI3L1]) is a secreted chitinase-like protein which is upregulated in cancers and suggested to have pro-tumorigenic activity. YKL-40 lacks enzymatic function, but it can bind carbohydrates such as chitin. Chitooligosaccharides (COS) derived from deacetylation and hydrolysis of chitin might be used for the blockade of YKL-40 function. Here, public single-cell RNA sequencing datasets were used to elucidate the cellular source of YKL-40 gene expression in human tumors. Fibroblasts and myeloid cells were the primary sources of YKL-40. Screening of YKL-40 gene expression in syngeneic mouse cancer models showed the highest expression in the Lewis lung carcinoma (LL2) model. LL2 was used to investigate COS monotherapy and combinations with immune checkpoint inhibitors (anti-PD-L1 and anti-CTLA-4) (ICIs) and radiotherapy (8 Gy × 3) (RT). COS tended to reduce plasma YKL-40 levels, but it did not affect tumor growth. LL2 showed minimal responses to ICIs, or to RT alone. Interestingly, ICIs combined with COS led to delayed tumor growth. RT also enhanced the efficacy of ICIs; however, the addition of COS did not further delay the tumor growth. COS may exert their anti-tumorigenic effects through the inhibition of YKL-40, but additional functions of COS should be investigated.
Collapse
|
22
|
Li X, Wang Y, Feng C, Chen H, Gao Y. Chemical Modification of Chitosan for Developing Cancer Nanotheranostics. Biomacromolecules 2022; 23:2197-2218. [PMID: 35522524 DOI: 10.1021/acs.biomac.2c00184] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer is a worldwide public health issue that has not been conquered. Theranostics, the combination of a therapeutic drug and imaging agent in one formulation using nanomaterials, has been developed to better cure cancer in recent years. Although diverse biomaterials have been applied in cancer theranostics, chitosan (CS), a natural polysaccharide bearing easy modification sites with excellent biocompatibility and biodegradability, shows great potential for developing cancer nanotheranostics. In this review, we seek to describe the chemical functionalities of CS used in cancer theranostics and their synthesis methods. We also present recent discoveries and research progresses on how the CS functionalization could improve the delivery efficiency of CS-based nanotheranostics. Finally, we report several case studies about the application of CS-based nanotheranostics. This paper focuses on the strategies to construct CS-based theranostics systems via chemical routes and highlights their applications in cancer treatment, which can provide useful references for further studies.
Collapse
Affiliation(s)
- Xudong Li
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou 350108, China
| | - Yuran Wang
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou 350108, China
| | - Chenyun Feng
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou 350108, China
| | - Haijun Chen
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Yu Gao
- Cancer Metastasis Alert and Prevention Center, College of Chemistry, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou 350108, China
| |
Collapse
|
23
|
Ding J, Guo Y. Recent Advances in Chitosan and its Derivatives in Cancer Treatment. Front Pharmacol 2022; 13:888740. [PMID: 35694245 PMCID: PMC9178414 DOI: 10.3389/fphar.2022.888740] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/13/2022] [Indexed: 12/15/2022] Open
Abstract
Cancer has become a main public health issue globally. The conventional treatment measures for cancer include surgery, radiotherapy and chemotherapy. Among the various available treatment measures, chemotherapy is still one of the most important treatments for most cancer patients. However, chemotherapy for most cancers still faces many problems associated with a lot of adverse effects, which limit its therapeutic potency, low survival quality and discount cancer prognosis. In order to decrease these side effects and improve treatment effectiveness and patient’s compliance, more targeted treatments are needed. Sustainable and controlled deliveries of drugs with controllable toxicities are expected to address these hurdles. Chitosan is the second most abundant natural polysaccharide, which has excellent biocompatibility and notable antitumor activity. Its biodegradability, biocompatibility, biodistribution, nontoxicity and immunogenicity free have made chitosan become a widely used polymer in the pharmacology, especially in oncotherapy. Here, we make a brief review of the main achievements in chitosan and its derivatives in pharmacology with a special focus on their agents delivery applications, immunomodulation, signal pathway modulation and antitumor activity to highlight their role in cancer treatment. Despite a large number of successful studies, the commercialization of chitosan copolymers is still a big challenge. The further development of polymerization technology may satisfy the unmet medical needs.
Collapse
Affiliation(s)
- Jingxian Ding
- Department of Radiation Oncology, The Breast Cancer Institute, The Third Hospital of Nanchang, Nanchang, China
| | - Yonghong Guo
- Department of Radiation Oncology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Yonghong Guo,
| |
Collapse
|
24
|
Mendez-Pfeiffer P, Juarez J, Hernandez J, Taboada P, Virués C, Valencia D, Velazquez C. Nanocarriers as drug delivery systems for propolis: A therapeutic approach. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102762] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
25
|
Chitosan: An Overview of Its Properties and Applications. Polymers (Basel) 2021; 13:polym13193256. [PMID: 34641071 PMCID: PMC8512059 DOI: 10.3390/polym13193256] [Citation(s) in RCA: 416] [Impact Index Per Article: 104.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/16/2021] [Accepted: 09/22/2021] [Indexed: 12/13/2022] Open
Abstract
Chitosan has garnered much interest due to its properties and possible applications. Every year the number of publications and patents based on this polymer increase. Chitosan exhibits poor solubility in neutral and basic media, limiting its use in such conditions. Another serious obstacle is directly related to its natural origin. Chitosan is not a single polymer with a defined structure but a family of molecules with differences in their composition, size, and monomer distribution. These properties have a fundamental effect on the biological and technological performance of the polymer. Moreover, some of the biological properties claimed are discrete. In this review, we discuss how chitosan chemistry can solve the problems related to its poor solubility and can boost the polymer properties. We focus on some of the main biological properties of chitosan and the relationship with the physicochemical properties of the polymer. Then, we review two polymer applications related to green processes: the use of chitosan in the green synthesis of metallic nanoparticles and its use as support for biocatalysts. Finally, we briefly describe how making use of the technological properties of chitosan makes it possible to develop a variety of systems for drug delivery.
Collapse
|
26
|
Deng S, Jin P, Sherchan P, Liu S, Cui Y, Huang L, Zhang JH, Gong Y, Tang J. Recombinant CCL17-dependent CCR4 activation alleviates neuroinflammation and neuronal apoptosis through the PI3K/AKT/Foxo1 signaling pathway after ICH in mice. J Neuroinflammation 2021; 18:62. [PMID: 33648537 PMCID: PMC7923481 DOI: 10.1186/s12974-021-02112-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/16/2021] [Indexed: 02/07/2023] Open
Abstract
Background Intracerebral hemorrhage (ICH), a devastating subtype of stroke, is associated with high mortality and morbidity. Neuroinflammation is an important factor leading to ICH-induced neurological injuries. C-C Chemokine Receptor 4 (CCR4) plays an important role in enhancing hematoma clearance after ICH. However, it is unclear whether CCR4 activation can ameliorate neuroinflammation and apoptosis of neurons following ICH. The aim of the present study was to examine the effects of recombinant CCL17 (rCCL17)-dependent CCR4 activation on neuroinflammation and neuronal apoptosis in an intrastriatal autologous blood injection ICH model, and to determine whether the PI3K/AKT/Foxo1 signaling pathway was involved. Methods Two hundred twenty-six adult (8-week-old) male CD1 mice were randomly assigned to sham and ICH surgery groups. An intrastriatal autologous blood injection ICH model was used. rCCL17, a CCR4 ligand, was delivered by intranasal administration at 1 h, 3 h, and 6 h post-ICH. CCL17 antibody was administrated by intraventricular injection at 1 h post-ICH. C021, a specific inhibitor of CCR4 and GDC0068, an AKT inhibitor were delivered intraperitoneally 1 h prior to ICH induction. Brain edema, neurobehavioral assessments, western blotting, Fluoro-Jade C staining, terminal deoxynucleotidyl transferase dUTP nick end labeling, and immunofluorescence staining were conducted. Results Endogenous expression of CCL17 and CCR4 were increased following ICH, peaking at 5 days post-induction. CCR4 was found to co-localize with microglia, neurons, and astrocytes. rCCL17 treatment decreased brain water content, attenuated short- and long-term neurological deficits, deceased activation of microglia/macrophages and infiltration of neutrophils, and inhibited neuronal apoptosis in the perihematomal region post-ICH. Moreover, rCCL17 treatment post-ICH significantly increased the expression of CCR4, PI3K, phosphorylated AKT, and Bcl-2, while Foxo1, IL-1β, TNF-α, and Bax expression were decreased. The neuroprotective effects of rCCL17 were reversed with the administration of C021 or GDC0068. Conclusions rCCL17-dependent CCR4 activation ameliorated neurological deficits, reduced brain edema, and ameliorated neuroinflammation and neuronal apoptosis, at least in part, through the PI3K/AKT/Foxo1 signaling pathway after ICH. Thus, activation of CCR4 may provide a promising therapeutic approach for the early management of ICH. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02112-3.
Collapse
Affiliation(s)
- Shuixiang Deng
- Department of Critical Care Medicine, HuaShan Hospital, Fudan University, 12 middle WuLuMuQi, Shanghai, 200040, China.,Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA
| | - Peng Jin
- Department of Critical Care Medicine, HuaShan Hospital, Fudan University, 12 middle WuLuMuQi, Shanghai, 200040, China.,Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA
| | - Prativa Sherchan
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA
| | - Shengpeng Liu
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA.,Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, Guangdong, China
| | - Yuhui Cui
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA
| | - Lei Huang
- Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA.,Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Ye Gong
- Department of Critical Care Medicine, HuaShan Hospital, Fudan University, 12 middle WuLuMuQi, Shanghai, 200040, China. .,Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Jiping Tang
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA.
| |
Collapse
|
27
|
Xia L, Oyang L, Lin J, Tan S, Han Y, Wu N, Yi P, Tang L, Pan Q, Rao S, Liang J, Tang Y, Su M, Luo X, Yang Y, Shi Y, Wang H, Zhou Y, Liao Q. The cancer metabolic reprogramming and immune response. Mol Cancer 2021; 20:28. [PMID: 33546704 PMCID: PMC7863491 DOI: 10.1186/s12943-021-01316-8] [Citation(s) in RCA: 557] [Impact Index Per Article: 139.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/15/2021] [Indexed: 02/07/2023] Open
Abstract
The overlapping metabolic reprogramming of cancer and immune cells is a putative determinant of the antitumor immune response in cancer. Increased evidence suggests that cancer metabolism not only plays a crucial role in cancer signaling for sustaining tumorigenesis and survival, but also has wider implications in the regulation of antitumor immune response through both the release of metabolites and affecting the expression of immune molecules, such as lactate, PGE2, arginine, etc. Actually, this energetic interplay between tumor and immune cells leads to metabolic competition in the tumor ecosystem, limiting nutrient availability and leading to microenvironmental acidosis, which hinders immune cell function. More interestingly, metabolic reprogramming is also indispensable in the process of maintaining self and body homeostasis by various types of immune cells. At present, more and more studies pointed out that immune cell would undergo metabolic reprogramming during the process of proliferation, differentiation, and execution of effector functions, which is essential to the immune response. Herein, we discuss how metabolic reprogramming of cancer cells and immune cells regulate antitumor immune response and the possible approaches to targeting metabolic pathways in the context of anticancer immunotherapy. We also describe hypothetical combination treatments between immunotherapy and metabolic intervening that could be used to better unleash the potential of anticancer therapies.
Collapse
Affiliation(s)
- Longzheng Xia
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Linda Oyang
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Jinguan Lin
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Shiming Tan
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Yaqian Han
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Nayiyuan Wu
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Pin Yi
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China.,University of South China, 421001, Hengyang, Hunan, China
| | - Lu Tang
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China.,University of South China, 421001, Hengyang, Hunan, China
| | - Qing Pan
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China.,University of South China, 421001, Hengyang, Hunan, China
| | - Shan Rao
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Jiaxin Liang
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Yanyan Tang
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Min Su
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Xia Luo
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Yiqing Yang
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Yingrui Shi
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Hui Wang
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China
| | - Yujuan Zhou
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China.
| | - Qianjin Liao
- Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, 410013, Changsha, Hunan, China.
| |
Collapse
|
28
|
Chen X, Zhou M, Fan W, Yang M, Yang L. Combination of Sodium Cantharidinate with Cisplatin Synergistically Hampers Growth of Cervical Cancer. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:171-183. [PMID: 33469269 PMCID: PMC7812528 DOI: 10.2147/dddt.s282777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/16/2020] [Indexed: 01/22/2023]
Abstract
Background Sodium cantharidinate (SC) has been broadly applied in lung cancer treatment in China, while its specific function in cervical cancer (CC), a great contributor to death of female reproductive system cancers, remains unclear. Our research evaluated the anti-tumor effects of SC in CC and the mechanism involved. Methods First, cisplatin (DDP)-resistant Caski-1 and ME180 cell lines were developed and treated with SC. The effects of SC on CC cell growth were then evaluated. Subsequently, the genes targeted by SC were predicted via the bioinformatics website. The correlations between PTPN1 expression and tumor stage, lymph node metastasis and tumor differentiation were examined. We further conducted rescue experiments by overexpressing PTPN1 in CC cells, followed by SC and cisplatin treatments. The activation of the PI3K/AKT pathway in CC cells, and the effect of SC on the growth and drug resistance of Caski-1 cells in vivo were investigated. Results The sensitivity of Caski-1 and ME180 cells to DDP was increased after SC treatment, which also enhanced the inhibitory effect of DDP on the cell growth. By prediction, we found that SC could target PTPN1. Patients with high expression of PTPN1 had higher clinical stage, lymph node metastasis and lower tumor differentiation. SC inhibited PTPN1 expression. Overexpression of PTPN1 attenuated the effect of SC. Furthermore, PTPN1 activated the PI3K/AKT pathway. Moreover, SC treatment inhibited the growth and drug resistance of Caski-1 cells in vivo. Conclusion SC promotes drug sensitivity of CC cells to DDP by targeting PTPN1, thereby impairing the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Xiangxun Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, People's Republic of China
| | - Mengxi Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, People's Republic of China
| | - Wenjie Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, People's Republic of China
| | - Mingwei Yang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, People's Republic of China
| | - Lin Yang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, People's Republic of China
| |
Collapse
|