1
|
Al Mashud MA, Devnath R, Anzuman M, Sumona MI, Hossain MS, Kumer A, Talukder MEK, Rahman MM, Imon RR, Akash S, El Moussaoui A, Salamatullah AM, Bourhia M. New Approach as Inhibitor Against Head-Neck Cancer by In silico, DFT, FMOs, Docking, Molecular Dynamic, and ADMET of Euphorbia tirucalli (Pencil Cactus). Med Chem 2025; 21:122-143. [PMID: 40007184 DOI: 10.2174/0115734064315601240628115330] [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: 03/10/2024] [Revised: 05/25/2024] [Accepted: 05/31/2024] [Indexed: 02/27/2025]
Abstract
BACKGROUND Head and neck cancer (HNC) is on the rise worldwide, endangering lives and straining healthcare systems in both developing and developed nations. Despite the availability of a number of therapy options, the success rate for treating and controlling head and neck cancer remains dismal. To combat the aggressiveness and drug resistance of Epstein-Barr virus (EBV)-positive Head-Neck cancer cells, this study looks into the potential of Euphorbia tirucalli (pencil cactus) leaf extract. OBJECTIVES The goal of this study is to identify prospective therapeutic candidates from the extract of Euphorbia tirucalli (pencil cactus) leaves, which have the ability to inhibit Epstein-Barr virus (EBV)-positive Head- Neck cancer cells. MATERIALS AND METHODS The thirteen most important chemical components found in Euphorbia tirucalli (pencil cactus) leaves were analyzed by means of molecular modeling techniques such as Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET), Quantum Mechanics (QM) calculation, molecular docking, and molecular dynamics (MD) simulations. Using the Prediction of Activity Spectra for Substances (PASS) model, we assess the potency of these compounds. Important molecular properties such as chemical potential, electronegativity, hardness, and softness can be determined with the use of quantum chemical calculations employing HOMO-LUMO analysis. These drugs' safety and toxicological characteristics are better understood to assessments of their pharmacokinetics and ADMET. Finally, molecular dynamics simulations are employed to verify binding interactions and assess the stability of docked complexes. RESULTS The molecular docking analysis identifies ligands (01), (02), and (10) as strong competitors, with strong binding affinity for the Epstein-Barr virus (EBV)-positive Head-Neck cancer cell line. Not only do the ligands (01), (02), and (10) match the criteria for a potential new inhibitor of head-neck cancer, but they also outperform the present FDA-approved treatment. CONCLUSION Taraxerol, euphol, and ephorginol, three phytochemicals isolated from the leaves of the Euphorbia tirucalli (pencil cactus), have been identified as effective anti-cancer agents with the potential to serve as a foundation for novel head-neck cancer therapies, particularly those targeting the Epstein-Barr virus (EBV)-overexpressing subtype of this disease. An effective, individualized treatment plan for head-neck cancer is a long way off, but this study is a major step forward that could change the lives of patients and reduce the global burden of this disease.
Collapse
Affiliation(s)
- Md Abdullah Al Mashud
- Biophysics and Biomedicine Research Lab, Department of Electrical and Electronic Engineering, Islamic University, Kushtia-7003, Bangladesh
- Computational Bio-info Lab, Research and Development Center for Sustainability, Scientific Foundation for Cancer Research, Kushtia-7000, Bangladesh
| | - Ramprosad Devnath
- Nanotechnology & Catalysis Research Centre (NANOCAT), Institute for Advanced Studies (IAS), Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Masuma Anzuman
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia-7003, Bangladesh
| | - Mahbuba Iasmin Sumona
- Biophysics and Biomedicine Research Lab, Department of Electrical and Electronic Engineering, Islamic University, Kushtia-7003, Bangladesh
| | - Md Shamim Hossain
- Department of Computer Science and Technology, University of Science and Technology of China, Hefei, Anhui, 230026, P.R. China
| | - Ajoy Kumer
- Laboratory of Computational Research for Drug Design and Material Science, Department of Chemistry, College of Arts and Sciences, IUBAT-International University of Business Agriculture and Technology, 4 Embankment Drive Road, Sector 10, Uttara Model Town, Dhaka 1230, Bangladesh
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Md Enamul Kabir Talukder
- Molecular and Cellular Biology Laboratory, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore-7408, Bangladesh
| | - Md Mashiar Rahman
- Molecular and Cellular Biology Laboratory, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore-7408, Bangladesh
| | - Raihan Rahman Imon
- Molecular and Cellular Biology Laboratory, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore-7408, Bangladesh
| | - Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Birulia 1216 Ashulia, Dhaka, Bangladesh
| | - Abdelfattah El Moussaoui
- Plant Biotechnology Team, Faculty of Sciences, Abdelmalek Essaadi University, Tetouan 93002, Morocco
| | - Ahmad Mohammad Salamatullah
- Department of Food Science & Nutrition, College of Food and Agricultural Sciences, King Saud University, 11 P.O. Box 2460, Riyadh 11451, Saudi Arabia
| | - Mohammed Bourhia
- Laboratory of Biotechnology and Natural Resources Valorization, Faculty of Sciences, Ibn Zohr University, 80060, Agadir, Morocco
| |
Collapse
|
2
|
Bishayee A, Penn A, Bhandari N, Petrovich R, DeLiberto LK, Burcher JT, Barbalho SM, Nagini S. Dietary plants for oral cancer prevention and therapy: A review of preclinical and clinical studies. Phytother Res 2024; 38:5225-5263. [PMID: 39193857 DOI: 10.1002/ptr.8293] [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: 01/13/2024] [Revised: 06/23/2024] [Accepted: 07/06/2024] [Indexed: 08/29/2024]
Abstract
Oral cancer is a disease with high mortality and rising incidence worldwide. Although fragmentary literature on the anti-oral cancer effects of plant products has been published, a comprehensive analysis is lacking. In this work, a critical and comprehensive evaluation of oral cancer preventative or therapeutic effects of dietary plants was conducted. An exhaustive analysis of available data supports that numerous dietary plants exert anticancer effects, including suppression of cell proliferation, viability, autophagy, angiogenesis, invasion, and metastasis while promoting cell cycle arrest and apoptosis. Plant extracts and products target several cellular mechanisms, such as the reversal of epithelial-to-mesenchymal transition and the promotion of oxidative stress and mitochondrial membrane dysfunction by modulation of various signaling pathways. These agents were also found to regulate cellular growth signaling pathways by action on extracellular signal-regulated kinase and mitogen-activated protein kinase, inflammation via modulation of cyclooxygenase (COX)-1, COX-2, and nuclear factor-κB p65, and metastasis through influence of cadherins and matrix metalloproteinases. In vivo studies support these findings and demonstrate a decrease in tumor burden, incidence, and hyperplastic and dysplastic changes. Clinical studies also showed decreased oral cancer risk. However, high-quality studies should be conducted to establish the clinical efficacy of these plants. Overall, our study supports the use of dietary plants, especially garlic, green tea, longan, peppermint, purple carrot, saffron, tomato, and turmeric, for oral cancer prevention and intervention. However, further research is required before clinical application of this strategy.
Collapse
Affiliation(s)
- Anupam Bishayee
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Amanda Penn
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Neha Bhandari
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Riley Petrovich
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Lindsay K DeLiberto
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Jack T Burcher
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Sandra Maria Barbalho
- School of Food and Technology of Marilia, Marília, São Paulo, Brazil
- School of Medicine, University of Marília, Marília, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília, Marília, Sao Paulo, Brazil
| | - Siddavaram Nagini
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalai Nagar, Tamil Nadu, India
| |
Collapse
|
3
|
Sun P, Gu KJ, Zheng G, Sikora AG, Li C, Zafereo M, Wei P, Wu J, Shete S, Liu J, Li G. Genetic variations associated with telomere length predict the risk of recurrence of non-oropharyngeal head and neck squamous cell carcinoma. Mol Carcinog 2024; 63:1722-1737. [PMID: 38837510 DOI: 10.1002/mc.23768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/15/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024]
Abstract
Genetic factors underlying lymphocyte telomere length (LTL) may provide insights into genomic stability and integrity, with direct links to susceptibility to cancer recurrence. Polymorphisms in telomere-associated genes are strongly associated with LTL and cancer risk, while few large studies have explored the associations between LTL-related polymorphisms and recurrence risk of non-oropharyngeal head and neck squamous cell carcinoma (non-OPHNSCC). Totally 1403 non-OPHNSCC patients were recruited and genotyped for 16 LTL-related polymorphisms identified by genome-wide association studies. Univariate and multivariate analyzes were performed to evaluate associations between the polymorphisms and non-OPHNSCC recurrence risk. Patients carrying rs755017 GA/GG, rs2487999 TC/TT, rs2736108 TC/TT, or rs6772228 AT/AA genotypes exhibited shorter DFS than those with the rs755017 AA, rs2487999 CC, rs2736108 CC, or s6772228 TT genotypes, respectively (all log-rank p < 0.05). Multivariable analysis confirmed an increased risk of recurrence for patients carrying rs755017 GA/GG, rs2487999 TC/TT, rs2736108 TC/TT, or rs6772228 AT/AA genotypes (adjusted hazard ratio [aHR]: 1.66, 95% confidence interval [CI]: 1.32-2.07; aHR: 1.77, 95% CI: 1.41-2.23; aHR: 1.56, 95% CI: 1.22-1.99; aHR: 1.52, 95% CI: 1.20-1.93, respectively). Further stratified analysis revealed stronger associations between these genotypes and recurrence risk in ever-smokers and patients undergoing chemoradiotherapy. The similar but particularly pronounced results were observed for the combined risk genotypes of the four significant polymorphisms. This is the first large study on non-OPHNSCC patients showing that LTL-related polymorphisms may modify risk of non-OPHNSCC recurrence individually and jointly, particularly when analyzed in the context of smoking status and personized treatment. Larger studies are needed to validate these results.
Collapse
Affiliation(s)
- Peng Sun
- Department of Otolaryngology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kyle J Gu
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Texas Tech University Health Sciences Center School of Medicine, Lubbock, Texas, USA
| | - Guibin Zheng
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Thyroid Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Andrew G Sikora
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Chao Li
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Head and Neck Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Mark Zafereo
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Peng Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jia Wu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sanjay Shete
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jisheng Liu
- Department of Otolaryngology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Guojun Li
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
4
|
Wang D, Woodcock E, Yang X, Nishikawa H, Sviderskaya EV, Oshima M, Edwards C, Zhang Y, Korchev Y. Exploration of individual colorectal cancer cell responses to H 2O 2 eustress using hopping probe scanning ion conductance microscopy. Sci Bull (Beijing) 2024; 69:1909-1919. [PMID: 38644130 DOI: 10.1016/j.scib.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 03/12/2024] [Accepted: 03/25/2024] [Indexed: 04/23/2024]
Abstract
Colorectal cancer (CRC), a widespread malignancy, is closely associated with tumor microenvironmental hydrogen peroxide (H2O2) levels. Some clinical trials targeting H2O2 for cancer treatment have revealed its paradoxical role as a promoter of cancer progression. Investigating the dynamics of cancer cell H2O2 eustress at the single-cell level is crucial. In this study, non-contact hopping probe mode scanning ion conductance microscopy (HPICM) with high-sensitive Pt-functionalized nanoelectrodes was employed to measure dynamic extracellular to intracellular H2O2 gradients in individual colorectal cancer Caco-2 cells. We explored the relationship between cellular mechanical properties and H2O2 gradients. Exposure to 0.1 or 1 mmol/L H2O2 eustress increased the extracellular to intracellular H2O2 gradient from 0.3 to 1.91 or 3.04, respectively. Notably, cellular F-actin-dependent stiffness increased at 0.1 mmol/L but decreased at 1 mmol/L H2O2 eustress. This H2O2-induced stiffness modulated AKT activation positively and glutathione peroxidase 2 (GPX2) expression negatively. Our findings unveil the failure of some H2O2-targeted therapies due to their ineffectiveness in generating H2O2, which instead acts eustress to promote cancer cell survival. This research also reveals the complex interplay between physical properties and biochemical signaling in cancer cells' antioxidant defense, illuminating the exploitation of H2O2 eustress for survival at the single-cell level. Inhibiting GPX and/or catalase (CAT) enhances the cytotoxic activity of H2O2 eustress against CRC cells, which holds significant promise for developing innovative therapies targeting cancer and other H2O2-related inflammatory diseases.
Collapse
Affiliation(s)
- Dong Wang
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa 920-1192, Japan
| | - Emily Woodcock
- Department of Medicine, Imperial College London, London W12 0NN, United Kingdom; Cell Biology Research Centre, Molecular and Clinical Sciences Research Institute, St George's, University of London, London SW17 0RE, United Kingdom
| | - Xi Yang
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa 920-1192, Japan
| | - Hiromi Nishikawa
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa 920-1192, Japan
| | - Elena V Sviderskaya
- Cell Biology Research Centre, Molecular and Clinical Sciences Research Institute, St George's, University of London, London SW17 0RE, United Kingdom
| | - Masanobu Oshima
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa 920-1192, Japan
| | - Christopher Edwards
- Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Yanjun Zhang
- WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa 920-1192, Japan; Department of Medicine, Imperial College London, London W12 0NN, United Kingdom.
| | - Yuri Korchev
- Department of Medicine, Imperial College London, London W12 0NN, United Kingdom; WPI Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kanazawa 920-1192, Japan.
| |
Collapse
|
5
|
Liu X, Miao R, Liu K, Xie Q, Zheng P, Zhu J, Zhang Y, Peng F. Panoramic analysis of cell death patterns reveals prognostic and immune profiles of head and neck squamous cell carcinoma. Am J Cancer Res 2024; 14:2584-2607. [PMID: 38859838 PMCID: PMC11162683 DOI: 10.62347/pmda6193] [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: 02/21/2024] [Accepted: 05/15/2024] [Indexed: 06/12/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) has been characterized by a low therapeutic response and poor prognosis. Currently, there are no reliable predictive models for HNSCC progression and therapeutic efficacy. This study explores the role of diverse patterns of cell death in tumor development, positing them as predictive factors of HNSCC prognosis. We utilized bulk transcriptome and single-cell transcriptome, align with clinical information from TCGA and GEO database, to analyze genes associated with 15 types of cell death and construct a cell death index (CDI) signature. The associations of CDI with tumor-infiltrating immune cells and immunotherapy-related biomarkers were also evaluated using various algorithms. The CDI signature emerged as a robust prognosis biomarker that could identify patients who can benefit potentially from immunotherapy, thus improving diagnostic accuracy and optimizing clinical decisions in HNSCC management. Notably, we discovered that CAAP1 deficiency not only induced apoptosis but also enhanced anti-tumor immunity, suggesting its potential as a target for clinical drug development.
Collapse
Affiliation(s)
- Xinyu Liu
- Department of Otolaryngology Head and Neck Surgery, Loudi Central HospitalLoudi 417011, Hunan, The People’s Republic of China
| | - Rui Miao
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South UniversityNo. 87 Xiangya Road, Changsha 410008, Hunan, The People’s Republic of China
- Otolaryngology Major Disease Research Key Laboratory of Hunan ProvinceNo. 87 Xiangya Road, Changsha 410008, Hunan, The People’s Republic of China
| | - Kui Liu
- Department of Otolaryngology Head and Neck Surgery, Loudi Central HospitalLoudi 417011, Hunan, The People’s Republic of China
| | - Qun Xie
- Department of Otolaryngology Head and Neck Surgery, Loudi Central HospitalLoudi 417011, Hunan, The People’s Republic of China
| | - Penghui Zheng
- Department of Otolaryngology Head and Neck Surgery, Loudi Central HospitalLoudi 417011, Hunan, The People’s Republic of China
| | - Junai Zhu
- Department of Gastroenterology, The Chinese University of Hong KongHong Kong SAR, China
| | - Ying Zhang
- Department of Otolaryngology Head and Neck Surgery, Loudi Central HospitalLoudi 417011, Hunan, The People’s Republic of China
| | - Fusen Peng
- Department of Otolaryngology Head and Neck Surgery, Loudi Central HospitalLoudi 417011, Hunan, The People’s Republic of China
| |
Collapse
|
6
|
Yang Y, Chen Y, Tang H, Zhang Z, Zhou X, Xu W. DTTZ suppresses ferroptosis and reverses mitochondrial dysfunction in normal tissues affected by chemotherapy. Biomed Pharmacother 2024; 172:116227. [PMID: 38335570 DOI: 10.1016/j.biopha.2024.116227] [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: 11/10/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Conventional antineoplastic therapies cause severe normal tissue damage and existing cytoprotectants with acute toxicities or potential tumor protection limit their clinical application. We evaluated the selective cytoprotection of 2,2-dimethylthiazolidine hydrochloride in this study, which could protect normal tissue toxicity without interfering antineoplastic therapies. By using diverse cell lines and A549 xenograft model, we discovered a synthetic aminothiol 2,2-dimethylthiazolidine hydrochloride selectively diminished normal cellular ferroptosis via SystemXc-/Glutathione Peroxidase 4 pathway upon antineoplastic therapies without interfering the anticancer efficacy. We revealed the malignant and non-malignant tissues presenting different energy metabolism patterns. And cisplatin induces disparate replicative stress, contributing to the distinguishable cytoprotection of 2,2-dimethylthiazolidine in normal and tumor cells. The compound pre-application could mitigate cisplatin-induced normal cellular mitochondrial oxidative phosphorylation (OXPHOS) dysfunction. Pharmacologic ablation of mitochondria reversed 2,2-dimethylthiazolidine chemoprotection against cisplatin in the normal cell line. Combined, these results provide a potential therapeutic adjuvant to selectively diminish normal tissue damages retaining antineoplastic efficacy.
Collapse
Affiliation(s)
- Yuwei Yang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Yuanfang Chen
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Haikang Tang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Ziqi Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Xiaoliang Zhou
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| | - Wenqing Xu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| |
Collapse
|
7
|
Rahman MM, Hossain MM, Islam S, Ahmed R, Majumder M, Dey S, Kawser M, Sarkar B, Himu MER, Chowdhury AA, Ahmed S, Biswas S, Anwar MM, Hussain MJ, Kumar Shil R, Baidya S, Parial R, Islam MM, Bharde A, Jayant S, Aland G, Khandare J, Uddin SB, Noman ASM. CTC together with Shh and Nrf2 are prospective diagnostic markers for HNSCC. BMC Mol Cell Biol 2024; 25:4. [PMID: 38336617 PMCID: PMC10858504 DOI: 10.1186/s12860-024-00500-0] [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: 12/04/2023] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND The lack of appropriate prognostic biomarkers remains a significant obstacle in the early detection of Head and Neck Squamous Cell Carcinoma (HNSCC), a cancer type with a high mortality rate. Despite considerable advancements in treatment, the success in diagnosing HNSCC at an early stage still needs to be improved. Nuclear factor erythroid 2-related factor 2 (Nrf2) and Sonic Hedgehog (Shh) are overexpressed in various cancers, including HNSCC, and have recently been proposed as possible therapeutic targets for HNSCC. Circulating Tumor Cell (CTC) is a novel concept used for the early detection of cancers, and studies have suggested that a higher CTC count is associated with the aggressiveness of HNSCC and poor survival rates. Therefore, we aimed to establish molecular markers for the early diagnosis of HNSCC considering Shh/Nrf2 overexpression in the background. In addition, the relation between Shh/Nrf2 and CTCs is still unexplored in HNSCC patients. METHODS In the present study, we selected a cohort of 151 HNSCC patients and categorized them as CTC positive or negative based on the presence or absence of CTCs in their peripheral blood. Data on demographic and clinicopathological features with the survival of the patients were analyzed to select the patient cohort to study Shh/Nrf2 expression. Shh and Nrf2 expression was measured by qRT-PCR. RESULTS Considering significant demographic [smoking, betel leaf (p-value < 0.0001)] and clinicopathological risk factors [RBC count (p < 0.05), Platelet count (p < 0.05), Neutrophil count (p < 0.005), MCV (p < 0.0001), NLR (p < 0.05), MLR (p < 0.05)], patients who tested positive for CTC also exhibited significant overexpression of Shh/Nrf2 in both blood and tissue compared to CTC-negative patients. A strong association exists between CTCs and tumor grade. Following chemotherapy (a combination of Cisplatin, 5FU, and Paclitaxel), the frequency of CTCs was significantly decreased in patients with HNSCC who had tested positive for CTCs. The Kaplan-Meier plot illustrated that a higher number of CTCs is associated with poorer overall survival (OS) in patients with HNSCC. CONCLUSIONS Detecting CTCs, and higher expression of Shh and Nrf2 in HNSCC patients' blood, can be a promising tool for diagnosing and prognosticating HNSCC.
Collapse
Affiliation(s)
- Md Mizanur Rahman
- Rangamati Medical College, Rangamati, Bangladesh
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, 4331, Bangladesh
- EuGEF Research Foundation, Chattogram, Bangladesh
| | - Muhammad Mosaraf Hossain
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, 4331, Bangladesh.
- EuGEF Research Foundation, Chattogram, Bangladesh.
| | - Shafiqul Islam
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, 4331, Bangladesh
- EuGEF Research Foundation, Chattogram, Bangladesh
- Present Address: Stem Cell Genetics, Institute of Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Ridwan Ahmed
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, 4331, Bangladesh
- EuGEF Research Foundation, Chattogram, Bangladesh
| | - Mohit Majumder
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, 4331, Bangladesh
- EuGEF Research Foundation, Chattogram, Bangladesh
| | - Shantu Dey
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, 4331, Bangladesh
- EuGEF Research Foundation, Chattogram, Bangladesh
| | - Md Kawser
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, 4331, Bangladesh
- EuGEF Research Foundation, Chattogram, Bangladesh
| | - Bishu Sarkar
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, 4331, Bangladesh
- EuGEF Research Foundation, Chattogram, Bangladesh
| | - Md Ejajur Rahman Himu
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, 4331, Bangladesh
- EuGEF Research Foundation, Chattogram, Bangladesh
| | - Ali Asgar Chowdhury
- Department of Radiotherapy, Chittagong Medical College, Chattogram, Bangladesh
| | - Shakera Ahmed
- Department of Surgery, Chittagong Medical College, Chattogram, Bangladesh
| | - Supran Biswas
- Department of Otolaryngology and Head Neck Surgery, Chittagong Medical College, Chattogram, Bangladesh
| | - Mostafa Mahfuzul Anwar
- Department of Otolaryngology and Head Neck Surgery, Chittagong Medical College, Chattogram, Bangladesh
| | - Mohammad Jamal Hussain
- Department of Otolaryngology and Head Neck Surgery, Rangamati Medical College, Rangamati, Bangladesh
| | - Rajib Kumar Shil
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, 4331, Bangladesh
- EuGEF Research Foundation, Chattogram, Bangladesh
| | - Sunanda Baidya
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, 4331, Bangladesh
- EuGEF Research Foundation, Chattogram, Bangladesh
| | - Ramendu Parial
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, 4331, Bangladesh
- EuGEF Research Foundation, Chattogram, Bangladesh
| | - Mohammed Moinul Islam
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, 4331, Bangladesh
- EuGEF Research Foundation, Chattogram, Bangladesh
| | - Atul Bharde
- Actorious Innovations and Research Pvt. Ltd., India and Simi Valley, Pune, CA, USA
| | - Sreeja Jayant
- Actorious Innovations and Research Pvt. Ltd., India and Simi Valley, Pune, CA, USA
| | - Gourishankar Aland
- Actorious Innovations and Research Pvt. Ltd., India and Simi Valley, Pune, CA, USA
| | - Jayant Khandare
- Actorious Innovations and Research Pvt. Ltd., India and Simi Valley, Pune, CA, USA
| | | | - Abu Shadat Mohammod Noman
- Department of Biochemistry & Molecular Biology, University of Chittagong, Chattogram, 4331, Bangladesh.
- EuGEF Research Foundation, Chattogram, Bangladesh.
| |
Collapse
|
8
|
Petkov N, Tadjer A, Encheva E, Cherkezova-Zheleva Z, Paneva D, Stoyanova R, Kukeva R, Dorkov P, Pantcheva I. Experimental and DFT Study of Monensinate and Salinomycinate Complexes Containing {Fe 3(µ 3-O)} 7+ Core. Molecules 2024; 29:364. [PMID: 38257278 PMCID: PMC10818969 DOI: 10.3390/molecules29020364] [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: 12/20/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Two trinuclear oxo-centred iron(III) coordination compounds of monensic and salinomycinic acids (HL) were synthesized and their spectral properties were studied using physicochemical/thermal methods (FT-IR, TG-DTA, TG-MS, EPR, Mössbauer spectroscopy, powder XRD) and elemental analysis. The data suggested the formation of [Fe3(µ3-O)L3(OH)4] and the probable complex structures were modelled using the DFT method. The computed spectral parameters of the optimized constructs were compared to the experimentally measured ones. In each complex, three metal centres were joined together at the axial position by a μ3-O unit to form a {Fe3O}7+ core. The antibiotics monoanions served as bidentate ligands through the carboxylate and hydroxyl groups located at the termini. The carboxylate moieties played a dual role bridging each two metal centres. Hydroxide anions secured the overall neutral character of the coordination species. Mössbauer spectra displayed asymmetric quadrupole doublets that were consistent with the existence of two types of high-spin iron(III) sites with different environments-two Fe[O5] and one Fe[O6] centres. The solid-state EPR studies confirmed the +3 oxidation state of iron with a total spin St = 5/2 per trinuclear cluster. The studied complexes are the first iron(III) coordination compounds of monensin and salinomycin reported so far.
Collapse
Affiliation(s)
- Nikolay Petkov
- Faculty of Chemistry and Pharmacy, Sofia University St. Kliment Ohridski, 1164 Sofia, Bulgaria; (A.T.); (E.E.)
| | - Alia Tadjer
- Faculty of Chemistry and Pharmacy, Sofia University St. Kliment Ohridski, 1164 Sofia, Bulgaria; (A.T.); (E.E.)
| | - Elzhana Encheva
- Faculty of Chemistry and Pharmacy, Sofia University St. Kliment Ohridski, 1164 Sofia, Bulgaria; (A.T.); (E.E.)
- Institute of Physical Chemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Zara Cherkezova-Zheleva
- Institute of Catalysis, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (Z.C.-Z.); (D.P.)
| | - Daniela Paneva
- Institute of Catalysis, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (Z.C.-Z.); (D.P.)
| | - Radostina Stoyanova
- Institute of General and Inorganic Chemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (R.S.); (R.K.)
| | - Rositsa Kukeva
- Institute of General and Inorganic Chemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (R.S.); (R.K.)
| | - Petar Dorkov
- Research and Development Department, Biovet Ltd., 4550 Peshtera, Bulgaria;
| | - Ivayla Pantcheva
- Faculty of Chemistry and Pharmacy, Sofia University St. Kliment Ohridski, 1164 Sofia, Bulgaria; (A.T.); (E.E.)
| |
Collapse
|
9
|
Yu H, Xue W, Yu H, Song Y, Liu X, Qin L, Wang S, Bao H, Gu H, Chen G, Zhao D, Tu Y, Cheng J, Wang L, Ai Z, Hu D, Wang L, Peng A. Single-cell transcriptomics reveals variations in monocytes and Tregs between gout flare and remission. JCI Insight 2023; 8:e171417. [PMID: 38063198 PMCID: PMC10795830 DOI: 10.1172/jci.insight.171417] [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: 04/12/2023] [Accepted: 10/25/2023] [Indexed: 12/18/2023] Open
Abstract
Gout commonly manifests as a painful, self-limiting inflammatory arthritis. Nevertheless, the understanding of the inflammatory and immune responses underlying gout flares and remission remains ambiguous. Here, based on single-cell RNA-Seq and an independent validation cohort, we identified the potential mechanism of gout flare, which likely involves the upregulation of HLA-DQA1+ nonclassical monocytes and is related to antigen processing and presentation. Furthermore, Tregs also play an essential role in the suppressive capacity during gout remission. Cell communication analysis suggested the existence of altered crosstalk between monocytes and other T cell types, such as Tregs. Moreover, we observed the systemic upregulation of inflammatory and cytokine genes, primarily in classical monocytes, during gout flares. All monocyte subtypes showed increased arachidonic acid metabolic activity along with upregulation of prostaglandin-endoperoxide synthase 2 (PTGS2). We also detected a decrease in blood arachidonic acid and an increase in leukotriene B4 levels during gout flares. In summary, our study illustrates the distinctive immune cell responses and systemic inflammation patterns that characterize the transition from gout flares to remission, and it suggests that blood monocyte subtypes and Tregs are potential intervention targets for preventing recurrent gout attacks and progression.
Collapse
Affiliation(s)
- Hanjie Yu
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Wen Xue
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Hanqing Yu
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Yaxiang Song
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Xinying Liu
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Ling Qin
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Shu Wang
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Hui Bao
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Hongchen Gu
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Guangqi Chen
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Dake Zhao
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Yang Tu
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Jiafen Cheng
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Liya Wang
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Zisheng Ai
- Department of Medical Statistics, Tongji University School of Medicine, Shanghai, China
| | - Dayong Hu
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Ling Wang
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| | - Ai Peng
- Center for Nephrology and Clinical Metabolomics and Division of Nephrology, Shanghai Tenth People’s Hospital, and
| |
Collapse
|
10
|
Li Z, Cai H, Zheng J, Chen X, Liu G, Lv Y, Ye H, Cai G. Mitochondrial-related genes markers that predict survival in patients with head and neck squamous cell carcinoma affect immunomodulation through hypoxia, glycolysis, and angiogenesis pathways. Aging (Albany NY) 2023; 15:10347-10369. [PMID: 37796226 PMCID: PMC10599748 DOI: 10.18632/aging.205081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 09/08/2023] [Indexed: 10/06/2023]
Abstract
Mitochondria play a crucial role in the occurrence and development of tumors. We used mitochondria-related genes for consistent clustering to identify three stable molecular subtypes of head and neck squamous cell carcinoma (HNSCC) with different prognoses, mutations, and immune characteristics. Significant differences were observed in clinical characteristics, immune microenvironment, immune cell infiltration, and immune cell scores. TP53 was the most significantly mutated; cell cycle-related pathways and tumorigenesis-related pathways were activated in different subtypes. Risk modeling was conducted using a multifactor stepwise regression method, and nine genes were identified as mitochondria-related genes affecting prognosis (DKK1, EFNB2, ITGA5, AREG, EPHX3, CHGB, P4HA1, CCND1, and JCHAIN). Risk score calculations revealed significant differences in prognosis, immune cell scores, immune cell infiltration, and responses to conventional chemotherapy drugs. Glycolysis, angiogenesis, hypoxia, and tumor-related pathways were positively correlated with the RiskScore. Clinical samples were subjected to qPCR to validate the results. In this work, we constructed a prognostic model based on the mitochondrial correlation score, which well reflects the risk and positive factors for the prognosis of patients with HNSCC. This model can be used to guide individualized adjuvant and immunotherapy in patients with HNSCC.
Collapse
Affiliation(s)
- Zhonghua Li
- Department of Otolaryngology Head and Neck Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, China
| | - Haoxi Cai
- School of Stomatology, Ningxia Medical University, Yinchuan 750004, China
| | - Jinyang Zheng
- Department of Pathology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, China
| | - Xun Chen
- Department of Oral Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou 362000, China
| | - Guancheng Liu
- Department of Otolaryngology Head and Neck Surgery, The Hospital Affiliated of Guilin Medical College, Guilin 541000, China
| | - Yunxia Lv
- Department of Thyroid Surgery, The Second Affiliated Hospital to Nanchang University, Nanchang 330006, China
| | - Hui Ye
- Haicang Hospital Affiliated of Xiamen Medical College, Xiamen 361026, China
| | - Gengming Cai
- Haicang Hospital Affiliated of Xiamen Medical College, Xiamen 361026, China
- The School of Clinical Medicine, Fujian Medical University, Fuzhou 361026, China
- The Graduate School of Fujian Medical University, Fuzhou 361026, China
| |
Collapse
|
11
|
Chang A, Wang Y, Guo X, Sun Z, Ling J, Pan J, Zhuo X. Identification of immune-related genes in the prognosis of head and neck cancer using a novel prognostic signature model. Oral Surg Oral Med Oral Pathol Oral Radiol 2023; 136:478-489. [PMID: 37620228 DOI: 10.1016/j.oooo.2023.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 06/11/2023] [Accepted: 07/02/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND Increasing evidence indicates that the immune response plays a critical role in the development of head and neck cancer (HNC). We aimed to develop an immune-related gene signature and evaluate its prognostic value in patients with HNC. METHODS We retrieved an HNC cohort from The Cancer Genome Atlas database and divided the samples into high-risk and low-risk groups based on the median of the immune and stromal scores. We performed Venn and Cox analyses to identify the immune-related DEGs to use in our prognostic model. We evaluated the correlation between the model and immune-cell infiltration and validated the prognostic value of the model by applying it to 2 external HNC cohorts. RESULTS We identified 7 DEGs-CCR4, WDFY4, VCAM1, LYZ, VSIG4, XIRP1, and CMKLR1-to use in our prognostic model and validated the model by applying it to 2 external HNC cohorts. We found that risk scores based on the model could reflect the status of the tumor microenvironment and that VSIG4 might be associated with lymph node metastasis in HNC. CONCLUSIONS We developed a highly accurate immune-related prognostic 7-gene model in HNC predication, indicating that these 7 genes play critical roles in the tumor microenvironment.
Collapse
Affiliation(s)
- Aoshuang Chang
- Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yan Wang
- Department of Respiratory Medicine, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xiaopeng Guo
- Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhen Sun
- Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou, China
| | - Junjun Ling
- Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jigang Pan
- School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xianlu Zhuo
- Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou, China.
| |
Collapse
|
12
|
Khedkar HN, Chen LC, Kuo YC, Wu ATH, Huang HS. Multi-Omics Identification of Genetic Alterations in Head and Neck Squamous Cell Carcinoma and Therapeutic Efficacy of HNC018 as a Novel Multi-Target Agent for c-MET/STAT3/AKT Signaling Axis. Int J Mol Sci 2023; 24:10247. [PMID: 37373393 DOI: 10.3390/ijms241210247] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Amongst the most prevalent malignancies worldwide, head and neck squamous cell carcinoma (HNSCC) is characterized by high morbidity and mortality. The failure of standard treatment modalities, such as surgery, radiotherapy, and chemotherapy, demands the need for in-depth understanding of the complex signaling networks involved in the development of treatment resistance. A tumor's invasive growth and high levels of intrinsic or acquired treatment resistance are the primary causes of treatment failure. This may be a result of the presence of HNSCC's cancer stem cells, which are known to have self-renewing capabilities that result in therapeutic resistance. Using bioinformatics methods, we discovered that elevated expressions of MET, STAT3, and AKT were associated with poor overall survival in HNSCC patients. We then evaluated the therapeutic potential of our newly synthesized small molecule HNC018 towards its potential as a novel anticancer drug. Our computer-aided structure characterization and target identification study predicted that HNC018 could target these oncogenic markers implicated in HNSCC. Subsequently, the HNC018 has demonstrated its anti-proliferative and anticancer activities towards the head and neck squamous cell carcinoma cell lines, along with displaying the stronger binding affinities towards the MET, STAT3, and AKT than the standard drug cisplatin. Reduction in the clonogenic and tumor-sphere-forming ability displays HNC018's role in decreasing the tumorigenicity. Importantly, an vivo study has shown a significant delay in tumor growth in HNC018 alone or in combination with cisplatin-treated xenograft mice model. Collectively with our findings, HNC018 highlights the desirable properties of a drug-like candidate and could be considered as a novel small molecule for treating head and neck squamous cell carcinoma.
Collapse
Affiliation(s)
- Harshita Nivrutti Khedkar
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, and Academia Sinica, Taipei 11031, Taiwan
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Lung-Ching Chen
- Division of Cardiology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 11101, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei 24205, Taiwan
| | - Yu-Cheng Kuo
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
| | - Alexander T H Wu
- Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Taipei Heart Institute (THI), Taipei Medical University, Taipei 11031, Taiwan
- Clinical Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Centre, Taipei 11490, Taiwan
| | - Hsu-Shan Huang
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, and Academia Sinica, Taipei 11031, Taiwan
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Centre, Taipei 11490, Taiwan
- School of Pharmacy, National Defense Medical Centre, Taipei 11490, Taiwan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
13
|
Budi HS, Farhood B. Targeting oral tumor microenvironment for effective therapy. Cancer Cell Int 2023; 23:101. [PMID: 37221555 DOI: 10.1186/s12935-023-02943-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/11/2023] [Indexed: 05/25/2023] Open
Abstract
Oral cancers are among the common head and neck malignancies. Different anticancer therapy modalities such as chemotherapy, immunotherapy, radiation therapy, and also targeted molecular therapy may be prescribed for targeting oral malignancies. Traditionally, it has been assumed that targeting malignant cells alone by anticancer modalities such as chemotherapy and radiotherapy suppresses tumor growth. In the last decade, a large number of experiments have confirmed the pivotal role of other cells and secreted molecules in the tumor microenvironment (TME) on tumor progression. Extracellular matrix and immunosuppressive cells such as tumor-associated macrophages, myeloid-derived suppressor cells (MDSCs), cancer-associated fibroblasts (CAFs), and regulatory T cells (Tregs) play key roles in the progression of tumors like oral cancers and resistance to therapy. On the other hand, infiltrated CD4 + and CD8 + T lymphocytes, and natural killer (NK) cells are key anti-tumor cells that suppress the proliferation of malignant cells. Modulation of extracellular matrix and immunosuppressive cells, and also stimulation of anticancer immunity have been suggested to treat oral malignancies more effectively. Furthermore, the administration of some adjuvants or combination therapy modalities may suppress oral malignancies more effectively. In this review, we discuss various interactions between oral cancer cells and TME. Furthermore, we also review the basic mechanisms within oral TME that may cause resistance to therapy. Potential targets and approaches for overcoming the resistance of oral cancers to various anticancer modalities will also be reviewed. The findings for targeting cells and potential therapeutic targets in clinical studies will also be reviewed.
Collapse
Affiliation(s)
- Hendrik Setia Budi
- Department of Oral Biology, Dental Pharmacology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
14
|
Utispan K, Koontongkaew S, Niyomtham N, Yingyongnarongkul BE. Ethanolic extract of Ocimum sanctum leaf modulates oxidative stress, cell cycle and apoptosis in head and neck cancer cell lines. Heliyon 2023; 9:e15518. [PMID: 37128326 PMCID: PMC10148043 DOI: 10.1016/j.heliyon.2023.e15518] [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: 12/14/2022] [Revised: 04/03/2023] [Accepted: 04/12/2023] [Indexed: 05/03/2023] Open
Abstract
Ocimum sanctum Linn. is a medicinal herb that has cytotoxic effects by inducing oxidative stress in some carcinomas. This study aimed to examine the impact of O. sanctum leaf extract on oxidative stress, cell cycle progression, and apoptosis in cell lines of head and neck squamous cell carcinoma (HNSCC). Isogenic primary (HN18/HN30) and metastatic (HN17/HN31) HNSCC cell lines were used. Preparation of the ethanolic extract of O. sanctum leaf (EEOS) was carried out. HNSCC cell lines were exposed to varying concentrations (0.1-0.8 mg/ml) of EEOS for a duration of 72 h, and the MTT assay was utilized to determine the cytotoxic doses. To assess the impact of EEOS on HNSCC cells, the levels of reactive oxygen species (ROS) and malondialdehyde were measured using a fluorometric method. Flow cytometry was utilized to evaluate effects of EEOS on the cell cycle, DNA damage, and apoptosis in HNSCC cells. Caspase-3 and -9 levels in the EEOS-treated HNSCC cells were measured by ELISA. The chemical components in EEOS were detected using high-performance liquid chromatography-electrospray ionization-time of flight-mass spectrometry. EEOS exhibited cytotoxicity against the HN18, HN17, HN30 and HN31 cells at minimum concentrations of 0.1, 0.3, 0.2 and 0.2 mg/ml, respectively. Treatment with EEOS resulted in a significant increase in ROS levels in HN18 and HN17 cells. Additionally, EEOS significantly induced the levels of malondialdehyde in HN18 and HN31 cells. Moreover, EEOS arrested the cell cycle in HN30 and HN31 cells, and significantly induced DNA damage and apoptosis in the HN18, HN30, and HN31 cells. EEOS selectively increased caspase-9 in the HN18 cells. However, caspase-3 was activated without apoptosis in the EEOS-treated HN17 cells. The constituents of EEOS were identified as rosmarinic acid, caffeic acid, and apigenin. In conclusion, EEOS exhibits various prooxidative and apoptotic effects between HNSCC cells.
Collapse
Affiliation(s)
- Kusumawadee Utispan
- Faculty of Dentistry, Thammasat University, Pathum Thani, 12120, Thailand
- Corresponding author.
| | - Sittichai Koontongkaew
- Walailak University International College of Dentistry, Walailak University, Bangkok, 10300, Thailand
| | - Nattisa Niyomtham
- Walailak University International College of Dentistry, Walailak University, Bangkok, 10300, Thailand
| | - Boon-ek Yingyongnarongkul
- Department of Chemistry and Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Ramkhamhaeng University, Bangkok, 10240, Thailand
| |
Collapse
|
15
|
Zhao L, Zhong B, Zhu Y, Zheng H, Wang X, Hou Y, Lu JJ, Ai N, Guo X, Ge W, Ma YY, Chen X. Nitrovin (difurazone), an antibacterial growth promoter, induces ROS-mediated paraptosis-like cell death by targeting thioredoxin reductase 1 (TrxR1). Biochem Pharmacol 2023; 210:115487. [PMID: 36893814 DOI: 10.1016/j.bcp.2023.115487] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/01/2023] [Accepted: 03/01/2023] [Indexed: 03/09/2023]
Abstract
Glioblastoma multiforme (GBM) is one of the most lethal malignant tumors in the human brain, with only a few chemotherapeutic drugs available after surgery. Nitrovin (difurazone) is widely used as an antibacterial growth promoter in livestock. Here, we reported that nitrovin might be a potential anticancer lead. Nitrovin showed significant cytotoxicity to a panel of cancer cell lines. Nitrovin induced cytoplasmic vacuolation, reactive oxygen species (ROS) generation, MAPK activation, and Alix inhibition but had no effect on caspase-3 cleavage and activity, suggesting paraptosis activation. Nitrovin-induced cell death of GBM cells was significantly reversed by cycloheximide (CHX), N-acetyl-l-cysteine (NAC), glutathione (GSH), and thioredoxin reductase 1 (TrxR1) overexpression. Vitamins C and E, inhibitors of pan-caspase, MAPKs, and endoplasmic reticulum (ER) stress failed to do so. Nitrovin-triggered cytoplasmic vacuolation was reversed by CHX, NAC, GSH, and TrxR1 overexpression but not by Alix overexpression. Furthermore, nitrovin interacted with TrxR1 and significantly inhibited its activity. In addition, nitrovin showed a significant anticancer effect in a zebrafish xenograft model, which was reversed by NAC. In conclusion, our results showed that nitrovin induced non-apoptotic and paraptosis-like cell death mediated by ROS through targeting TrxR1. Nitrovin might be a promising anticancer lead for further development.
Collapse
Affiliation(s)
- Lin Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Bingling Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Yanyan Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Haoyi Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Xumei Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Ying Hou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Nana Ai
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Macao, China
| | - Xiuli Guo
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Wei Ge
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Macao, China
| | - Yan-Yan Ma
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao, China.
| |
Collapse
|
16
|
Naghaviyan A, Hashemi-Moghaddam H, Zavareh S, Ebrahimi Verkiani M, Meuller A. Synergistic Effect Evaluation of Magnetotherapy and a Cationic-Magnetic Nanocomposite Loaded with Doxorubicin for Targeted Drug Delivery to Breast Adenocarcinoma. Mol Pharm 2023; 20:101-117. [PMID: 36475680 DOI: 10.1021/acs.molpharmaceut.2c00505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This work investigates the synergistic effect of magnetotherapy and a novel cationic-magnetic drug delivery system on inhibiting breast cancer cell growth and other tissues. First, super-paramagnetic magnetite (Fe3O4) nanoparticles were coated with doxorubicin-imprinted poly(methacrylic acid-co-diallyl dimethylammonium chloride) [Fe3O4/poly(MAA-DDA)]. The cationic-magnetic nanocomposite (CMC) was characterized using XRD, FT-IR, VSM, TGA, TEM, FESEM, EDS, DLS, and BET. In vitro analyses, including drug release kinetics, cytotoxicity, and hemolytic assays, confirmed this novel CMC's good drug release profile and biocompatibility. Finally, in vivo experiments on BALB/c mice were designed to evaluate the synergistic effect of magnetotherapy on targeted drug delivery using the CMC. In vivo fluorescence imaging evaluated the drug distribution in different tissues of mice. Tumor volume evaluation demonstrated the efficiency of the CMC and magnetotherapy in preventing tumor growth; the two techniques significantly reduced tumor volume. Histopathological analysis proved that applying magnetotherapy in conjunction with the cationic-magnetic drug delivery system significantly prevented tumor cell proliferation and increased apoptosis with limited impact on other tissues. Also, Dox and Fe concentrations in different tissues confirmed the efficient drug delivery to tumor cells.
Collapse
Affiliation(s)
- Alireza Naghaviyan
- Department of Pharmacy, Damghan Branch, Islamic Azad University, 3671637849Damghan, Iran
| | | | - Saeed Zavareh
- School of Biology, Damghan University, 3671641167Damghan, Iran
| | | | - Anja Meuller
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, Michigan48859, United States
| |
Collapse
|
17
|
Intratumoral pro-oxidants promote cancer immunotherapy by recruiting and reprogramming neutrophils to eliminate tumors. Cancer Immunol Immunother 2023; 72:527-542. [PMID: 36066649 PMCID: PMC9446783 DOI: 10.1007/s00262-022-03248-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 06/23/2022] [Indexed: 11/06/2022]
Abstract
Neutrophils have recently gained recognition for their potential in the fight against cancer. Neutrophil plasticity between the N1 anti-tumor and N2 pro-tumor subtypes is now apparent, as is the ability to polarize these individual subtypes by interventions such as intratumoral injection of various agents including bacterial products or pro-oxidants. Metabolic responses and the production of reactive oxygen species (ROS) such as hydrogen peroxide act as potent chemoattractants and activators of N1 neutrophils that facilitates their recruitment and ensuing activation of a toxic respiratory burst in tumors. Greater understanding of the precise mechanism of N1 neutrophil activation, recruitment and regulation is now needed to fully exploit their anti-tumor potential against cancers both locally and at distant sites. This systematic review critically analyzes these new developments in cancer immunotherapy.
Collapse
|
18
|
Ashrafi F, Rahimzada M, Parandi M, Mirhosseini A, Mashkani B, Ahmadi Ghezeldasht S, Soltani A, Rafatpanah H, Mosavat A, Abdolrahim Rezaee S. Molecular insight into the study of adult T-cell leukemia/lymphoma (ATLL): Ten-year studies on HTLV-1 associated diseases in an endemic region. Gene 2022; 847:146885. [PMID: 36108787 DOI: 10.1016/j.gene.2022.146885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/30/2022] [Accepted: 09/07/2022] [Indexed: 11/18/2022]
Abstract
The outcome of successful infection, including human T-cell leukemia virus type 1 (HTLV-1), is determined by the interactions between the host and the infectious agent. Ten years of work on HTLV-1-associated diseases in an endemic region of Iran have been critically compared in the present study. The outstanding findings of RNA-seq, system biology analysis, and gene expression measurements on adult T-cell leukemia/lymphoma (ATLL) and enzootic bovine leukosis(EBL) in our lab encouraged us to investigate the significant role of oncogenes in the ATLL malignancy. Most studies assessed such interactions by the proviral load (PVL), Tax, and HBZ regulatory proteins in HTLV-1 and the host's immunological and cell cycle factors. The current study is a comprehensive comparing view of our previously published and unpublished results investigating the HTLV-1-host interactions leading to the transformation of the infected cell. The main focus has been on the essential proteins implicated in the virus dissemination, cell survival, and proliferation of infected cells toward leukemia development and progression. Similar to its homolog BLV-AS-1-2 in EBL, the HTLV-1-HBZ is a pivotal factor in the maintenance and progression of the ATLL. In addition, the inappropriate activities of the PI3K/Akt pathway, BRCAs, and RAD51 in the DNA repair system, which are orchestrating many other immortalization pathways, might be the central factors in the manifestation of ATLL. HTLV-1-HBZ and the host PI3K/Akt pathway, BCAs, and RAD51 could be suggested as influential targets for the prognosis and proper therapy of ATLL.
Collapse
Affiliation(s)
- Fereshteh Ashrafi
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Masooma Rahimzada
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahsa Parandi
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Mirhosseini
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Baratali Mashkani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Sanaz Ahmadi Ghezeldasht
- Blood Borne Infections Research Center, Academic Center for Education, Culture, and, Research (ACECR), Razavi Khorasan, Mashhad, Iran.
| | - Ararsh Soltani
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Houshang Rafatpanah
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran; HTLV-1 Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Arman Mosavat
- Blood Borne Infections Research Center, Academic Center for Education, Culture, and, Research (ACECR), Razavi Khorasan, Mashhad, Iran.
| | - Seyed Abdolrahim Rezaee
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran; HTLV-1 Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
19
|
Liu K, Huang T, Zhang H, Deng H, Tang M. Establishment and validation of a redox-related long non-coding RNAs prognostic signature in head and neck squamous cell carcinoma. Sci Rep 2022; 12:22040. [PMID: 36543836 PMCID: PMC9772388 DOI: 10.1038/s41598-022-26490-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Reduction and oxidation (redox) reactions occur in living organisms as part of normal cellular metabolism. Here, we established a novel redox-related long non-coding RNAs (rrlncRNAs) signature to predict the prognosis and therapeutic response in Head and neck squamous cell carcinoma (HNSCC). The expression profile and clinical information were obtained from the TCGA project. In total, 10 differently expressed rrlncRNAs associated with prognosis were identified and involved in a prognostic risk score signature by the least absolute shrinkage and selection operator penalized Cox analysis. The area under the receiver operating characteristic curves of the survival rates predicted by the rrlncRNAs signature over one, two, and three years were found to be 0.651, 0.670, and 0.679. Following the completion of the Kaplan-Meier survival study, we discovered that the lower-risk cohort exhibited a much longer overall survival period in contrast with the higher-risk cohort. Univariate and multivariable Cox regression analyses demonstrated that the risk score independently served as a significant predictive factor. GO annotation and KEGG pathway analyses illustrated that the rrlncRNAs signature was strongly associated with immune-related functions as well as signaling pathways. The tumor-infiltrating immune cells, tumor microenvironment, immune-related functions, HLA gene family expression, immune checkpoint genes expression, and somatic variants differed substantially between the low- and high-risk cohorts. Moreover, patients in low-risk group were predicted to present a favorable immunotherapy responsiveness, while in contrast, the high-risk group patients might have a stronger sensitivity to "docetaxel". According to our findings, the rrlncRNAs signature showed an excellent prognosis predictive value and might indicate therapeutic response to immunotherapy in HNSCC.
Collapse
Affiliation(s)
- Kaitai Liu
- grid.507012.10000 0004 1798 304XDepartment of Radiation Oncology, The Lihuili Hospital, Ningbo Medical Center, Ningbo, Zhejiang China
| | - Tianyi Huang
- grid.507012.10000 0004 1798 304XDepartment of Radiation Oncology, The Lihuili Hospital, Ningbo Medical Center, Ningbo, Zhejiang China
| | - Hui Zhang
- grid.507012.10000 0004 1798 304XDepartment of Radiation Oncology, The Lihuili Hospital, Ningbo Medical Center, Ningbo, Zhejiang China
| | - Hongxia Deng
- Department of Otorhinolaryngology Head and Neck Surgery, The Lihuili Hospital, Ningbo Medical Center, Ningbo, Zhejiang China
| | - Ming Tang
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Women and Children’s Hospital, Ningbo, Zhejiang China
| |
Collapse
|
20
|
Bae I, Kim TG, Kim T, Kim D, Kim DH, Jo J, Lee YJ, Jeong YI. Phenethyl Isothiocyanate-Conjugated Chitosan Oligosaccharide Nanophotosensitizers for Photodynamic Treatment of Human Cancer Cells. Int J Mol Sci 2022; 23:13802. [PMID: 36430279 PMCID: PMC9693342 DOI: 10.3390/ijms232213802] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/02/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
The aim of this study is to synthesize phenethyl-conjugated chitosan oligosaccharide (COS) (abbreviated as ChitoPEITC) conjugates and then fabricate chlorin E6 (Ce6)-incorporated nanophotosensitizers for photodynamic therapy (PDT) of HCT-116 colon carcinoma cells. PEITC was conjugated with the amine group of COS. Ce6-incorporated nanophotosensitizers using ChitoPEITC (ChitoPEITC nanophotosensitizers) were fabricated by dialysis method. 1H nuclear magnetic resonance (NMR) spectra showed that specific peaks of COS and PEITC were observed at ChitoPEITC conjugates. Transmission electron microscope (TEM) confirmed that ChitoPEITC nanophotosensitizers have spherical shapes with small hydrodynamic diameters less than 200 nm. The higher PEITC contents in the ChitoPEITC copolymer resulted in a slower release rate of Ce6 from nanophotosensitizers. Furthermore, the higher Ce6 contents resulted in a slower release rate of Ce6. In cell culture study, ChitoPEITC nanophotosensitizers showed low toxicity against normal CCD986Sk human skin fibroblast cells and HCT-116 human colon carcinoma cells in the absence of light irradiation. ChitoPEITC nanophotosensitizers showed a significantly higher Ce6 uptake ratio than that of free Ce6. Under light irradiation, cellular reactive oxygen species (ROS) production of nanophotosensitizers was significantly higher than that of free Ce6. Especially, PEITC and/or ChitoPEITC themselves contributed to the production of cellular ROS regardless of light irradiation. ChitoPEITC nanophotosensitizers showed significantly higher PDT efficacy against HCT-116 cells than that of free Ce6. These results indicate that ChitoPEITC nanophotosensitizers have superior potential in Ce6 uptake, ROS production and PDT efficacy. In the HCT-116 cell-bearing mice tumor-xenograft model, ChitoPEITC nanophotosensitizers efficiently inhibited growth of tumor volume rather than free Ce6. In the animal imaging study, ChitoPEITC nanophotosensitizers were concentrated in the tumor tissue, i.e., fluorescence intensity in the tumor tissue was stronger than that of other tissues. We suggest that ChitoPEITC nanophotosensitizers are a promising candidate for the treatment of human colon cancer cells.
Collapse
Affiliation(s)
- Inho Bae
- Department of Dental Materials, College of Dentistry, Chosun University, Gwangju 61452, Korea
| | - Taeyu Grace Kim
- Tyros Biotechnology Inc., 75 Kneeland St. 14 floors, Boston, MA 02111, USA
- Brookline High School, 115 Greenough St., Brookline, MA 02445, USA
| | - Taeyeon Kim
- College of Art & Science, University of Pennsylvania, 249 S 36th St., Philadelphia, PA 19104, USA
| | - Dohoon Kim
- Tyros Biotechnology Inc., 75 Kneeland St. 14 floors, Boston, MA 02111, USA
| | - Doug-Hoon Kim
- Department of Optometry, Masan University, Changwon 51217, Korea
| | - Jaewon Jo
- Gwangju Center, Korea Basic Science Institute, Gwangju 61186, Korea
| | - Young-Ju Lee
- Gwangju Center, Korea Basic Science Institute, Gwangju 61186, Korea
| | - Young-Il Jeong
- Tyros Biotechnology Inc., 75 Kneeland St. 14 floors, Boston, MA 02111, USA
| |
Collapse
|
21
|
Li R, Zhao W, Wu T, Wang A, Li Q, Liu Y, Xiong H. Tantalum-carbon-integrated nanozymes as a nano-radiosensitizer for radiotherapy enhancement. Front Bioeng Biotechnol 2022; 10:1042646. [PMID: 36353740 PMCID: PMC9638097 DOI: 10.3389/fbioe.2022.1042646] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/12/2022] [Indexed: 04/16/2025] Open
Abstract
Radiotherapy (RT) plays a pivotal role in the comprehensive treatment of multiple malignant tumors, exerting its anti-tumor effects through direct induction of double-strand breaks (DSBs) or indirect induction of reactive oxygen species (ROS) production. However, RT resistance remains a therapeutic obstacle that leads to cancer recurrence and treatment failure. In this study, we synthesised a tantalum-carbon-integrated nanozyme with excellent catalase-like (CAT-like) activity and radiosensitivity by immobilising an ultrasmall tantalum nanozyme into a metal-organic framework (MOF)-derived carbon nanozyme through in situ reduction. The integrated tantalum nanozyme significantly increased the CAT activity of the carbon nanozyme, which promoted the production of more oxygen and increased the ROS levels. By improving hypoxia and increasing the level of ROS, more DNA DSBs occur at the cellular level, which, in turn, improves the sensitivity of RT. Moreover, tantalum-carbon-integrated nanozymes combined with RT have demonstrated notable anti-tumor activity in vivo. Therefore, exploiting the enzymatic activity and the effect of ROS amplification of this nanozyme has the potential to overcome resistance to RT, which may offer new horizons for nanozyme-based remedies for biomedical applications.
Collapse
Affiliation(s)
- Rui Li
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Weiheng Zhao
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Wu
- Department of Pharmacy, Henan Provincial People’s Hospital, Department of Pharmacy of Centeral China Fuwai Hospital, Centeral China Fuwai Hospital of Zhengzhou University, Zheng Zhou, China
| | - Aifeng Wang
- Department of Pharmacy, Henan Provincial People’s Hospital, Department of Pharmacy of Centeral China Fuwai Hospital, Centeral China Fuwai Hospital of Zhengzhou University, Zheng Zhou, China
| | - Qing Li
- Department of Pharmacy, Henan Provincial People’s Hospital, Department of Pharmacy of Centeral China Fuwai Hospital, Centeral China Fuwai Hospital of Zhengzhou University, Zheng Zhou, China
| | - Ying Liu
- Department of Pharmacy, Henan Provincial People’s Hospital, Department of Pharmacy of Centeral China Fuwai Hospital, Centeral China Fuwai Hospital of Zhengzhou University, Zheng Zhou, China
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Hong SO, Kook MS, Jeong YIL, Park MJ, Yang SW, Kim BH. Nanophotosensitizers Composed of Phenyl Boronic Acid Pinacol Ester-Conjugated Chitosan Oligosaccharide via Thioketal Linker for Reactive Oxygen Species-Sensitive Delivery of Chlorin e6 against Oral Cancer Cells. MATERIALS (BASEL, SWITZERLAND) 2022; 15:7057. [PMID: 36295132 PMCID: PMC9604738 DOI: 10.3390/ma15207057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/23/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2023]
Abstract
Chlorin E6 (Ce6)-incorporated nanophotosensitizers were fabricated for application in photodynamic therapy (PDT) of oral cancer cells. For this purpose, chitosan oligosaccharide (COS) was conjugated with hydrophobic and reactive oxygen species (ROS)-sensitive moieties, such as phenyl boronic acid pinacol ester (PBAP) via a thioketal linker (COSthPBAP). ThdCOOH was conjugated with PBAP to produce ThdCOOH-PBAP conjugates and then attached to amine groups of COS to produce a COSthPBAP copolymer. Ce6-incorporated nanophotosensitizers using the COSthPBAP copolymer were fabricated through the nanoprecipitation and dialysis methods. The Ce6-incorporated COSthPBAP nanophotosensitizers had a small diameter of less than 200 nm with a mono-modal distribution pattern. However, it became a multimodal and/or irregular distribution pattern when H2O2 was added. In a morphological observation using TEM, the nanophotosensitizers were disintegrated by the addition of H2O2, indicating that the COSthPBAP nanophotosensitizers had ROS sensitivity. In addition, the Ce6 release rate from the COSthPBAP nanophotosensitizers accelerated in the presence of H2O2. The SO generation was also higher in the nanophotosensitizers than in the free Ce6. Furthermore, the COSthPBAP nanophotosensitizers showed a higher intracellular Ce6 uptake ratio and ROS generation in all types of oral cancer cells. They efficiently inhibited the viability of oral cancer cells under light irradiation, but they did not significantly affect the viability of either normal cells or cancer cells in the absence of light irradiation. The COSthPBAP nanophotosensitizers showed a tumor-specific delivery capacity and fluorescence imaging of KB tumors in an in vivo animal tumor imaging study. We suggest that COSthPBAP nanophotosensitizers are promising candidates for the imaging and treatment of oral cancers.
Collapse
Affiliation(s)
- Sung-Ok Hong
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Kyung Hee University, Seoul 02447, Korea
- Department of Oral and Maxillofacial Surgery, Kyung Hee University Dental Hospital at Gangdong, Seoul 05278, Korea
| | - Min-Suk Kook
- Department of Maxillofacial Oral Surgery, School of Dentistry, Chonnam National University, Gwangju 61186, Korea
| | - Young-IL Jeong
- Department of Dental Materials, College of Dentistry, Chosun University, Gwangju 61452, Korea
| | - Min-Ju Park
- Department of Dental Materials, College of Dentistry, Chosun University, Gwangju 61452, Korea
| | - Seong-Won Yang
- Department of Ophthalmology, College of Medicine, Chosun University, Gwangju 61453, Korea
| | - Byung-Hoon Kim
- Department of Dental Materials, College of Dentistry, Chosun University, Gwangju 61452, Korea
| |
Collapse
|
23
|
Gao F, Gao J, Wang K, Song L. Efficacy and safety of transarterial chemoembolization with CalliSpheres® Microspheres in head and neck cancer. Front Surg 2022; 9:938305. [PMID: 36090318 PMCID: PMC9452835 DOI: 10.3389/fsurg.2022.938305] [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: 05/07/2022] [Accepted: 07/19/2022] [Indexed: 12/24/2022] Open
Abstract
Objective Transarterial chemoembolization with CalliSpheres® Microspheres (CSM-TACE) presents favorable efficacy and tolerable safety in several cancers, while its application in head and neck cancer (HNC) is unclear. Thus, the current pilot study aims to evaluate the efficacy and safety of CSM-TACE in treating HNC. Methods A total of 15 HNC patients receiving CSM-TACE at the Second Affiliated Hospital of Dalian Medical University from March 2017 to December 2021 were enrolled in this study. Moreover, treatment information, treatment response, progression-free survival (PFS), overall survival (OS), changes in liver and renal function indices, and adverse events were recorded. Results There were nine patients receiving CSM-TACE as first-line treatment and six patients receiving CSM-TACE as second- or above-line treatment; meanwhile, there were seven, seven, and one patient undergoing one time, two times, and three times of CSM-TACE, respectively. Furthermore, the objective response rate (ORR) and the disease control rate (DCR) were 60.0% and 100%, respectively, at the first month; meanwhile, the ORR and the DCR were 53.3% and 73.3%, respectively, at the second month. Moreover, the 1-year PFS rate was 34.1%, and the 1-year OS rate was 38.9%. Additionally, no change in liver function indices (namely, total protein, albumin, total bilirubin, alanine aminotransferase, and aspartate aminotransferase) or in renal function indices (namely, creatinine and blood urea nitrogen) was found before and 1 month after treatment (all P > 0.05). Meanwhile, no severe adverse events were found during and after CSM-TACE. Conclusion CSM-TACE illustrates favorable treatment response and survival benefits as well as a tolerable safety profile in HNC patients.
Collapse
Affiliation(s)
| | | | | | - Lei Song
- Correspondence: Lei Song Kuiyang Wang
| |
Collapse
|
24
|
Pan Z, Dong H, Huang N, Fang J. Oxidative stress and inflammation regulation of sirtuins: New insights into common oral diseases. Front Physiol 2022; 13:953078. [PMID: 36060706 PMCID: PMC9437461 DOI: 10.3389/fphys.2022.953078] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/29/2022] [Indexed: 12/22/2022] Open
Abstract
Sirtuins are a family of nicotinamide adenine dinucleotide (NAD)+-dependent histone deacetylases, comprising seven members SIRT1-SIRT7. Sirtuins have been extensively studied in regulating ageing and age-related diseases. Sirtuins are also pivotal modulators in oxidative stress and inflammation, as they can regulate the expression and activation of downstream transcriptional factors (such as Forkhead box protein O3 (FOXO3a), nuclear factor erythroid 2-related factor 2 (Nrf2) and nuclear factor-kappa B (NF-κB)) as well as antioxidant enzymes, through epigenetic modification and post-translational modification. Most importantly, studies have shown that aberrant sirtuins are involved in the pathogenesis of infectious and inflammatory oral diseases, and oral cancer. In this review, we provide a comprehensive overview of the regulatory patterns of sirtuins at multiple levels, and the essential roles of sirtuins in regulating inflammation, oxidative stress, and bone metabolism. We summarize the involvement of sirtuins in several oral diseases such as periodontitis, apical periodontitis, pulpitis, oral candidiasis, oral herpesvirus infections, dental fluorosis, and oral cancer. At last, we discuss the potential utilization of sirtuins as therapeutic targets in oral diseases.
Collapse
Affiliation(s)
- Zijian Pan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hao Dong
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ning Huang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jie Fang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jie Fang,
| |
Collapse
|
25
|
Siddique S, Chow JCL. Recent Advances in Functionalized Nanoparticles in Cancer Theranostics. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2826. [PMID: 36014691 PMCID: PMC9416120 DOI: 10.3390/nano12162826] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/10/2022] [Accepted: 08/16/2022] [Indexed: 05/21/2023]
Abstract
Cancer theranostics is the combination of diagnosis and therapeutic approaches for cancer, which is essential in personalized cancer treatment. The aims of the theranostics application of nanoparticles in cancer detection and therapy are to reduce delays in treatment and hence improve patient care. Recently, it has been found that the functionalization of nanoparticles can improve the efficiency, performance, specificity and sensitivity of the structure, and increase stability in the body and acidic environment. Moreover, functionalized nanoparticles have been found to possess a remarkable theranostic ability and have revolutionized cancer treatment. Each cancer treatment modality, such as MRI-guided gene therapy, MRI-guided thermal therapy, magnetic hyperthermia treatment, MRI-guided chemotherapy, immunotherapy, photothermal and photodynamic therapy, has its strengths and weaknesses, and combining modalities allows for a better platform for improved cancer control. This is why cancer theranostics have been investigated thoroughly in recent years and enabled by functionalized nanoparticles. In this topical review, we look at the recent advances in cancer theranostics using functionalized nanoparticles. Through understanding and updating the development of nanoparticle-based cancer theranostics, we find out the future challenges and perspectives in this novel type of cancer treatment.
Collapse
Affiliation(s)
- Sarkar Siddique
- Department of Physics, Toronto Metropolitan University, Toronto, ON M5B 2K3, Canada
| | - James C L Chow
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1X6, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON M5T 1P5, Canada
| |
Collapse
|
26
|
Xie W, Ye J, Guo Z, Lu J, Gao X, Wei Y, Zhao L. Ultrafast Fabrication of Iron/Manganese Co-Doped Bismuth Trimetallic Nanoparticles: A Thermally Aided Chemodynamic/Radio-Nanoplatform for Low-Dose Radioresistance. ACS APPLIED MATERIALS & INTERFACES 2022; 14:21931-21944. [PMID: 35511491 DOI: 10.1021/acsami.2c02484] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Low-dose radioresistance continues to be one of the major limitations for clinical curative treatment of cancer. Luckily, nanotechnology mediated by multifunctional nanomaterials provides potential opportunity to relieve the radioresistance via increasing the radiosensitivity of cancer cells. Herein, an ultrafast fabrication strategy is reported to prepare iron/manganese co-doped bismuth trimetallic nanoparticles (pFMBi NPs) as a multifunctional radiosensitizer for combined therapy. The bismuth matrix provides the intrinsic radiosensitization effect under the low and safe radiation dose via Auger electrons, photoelectrons, and Rayleigh scattering. Meanwhile, co-doping of iron and manganese ions endows pFMBi NPs with both the Fenton reaction property for reactive oxygen species (ROS) generation and photothermal conversion performance for heat production. Additional ROS generation enhances the radiosensitization effect by collaborating with Rayleigh scattering-mediated water radiolysis, and endogenous heat production under near-infrared 808 nm laser irradiation makes DNA more sensitive to radiation and ROS damage. Both in vitro and in vivo evaluations demonstrate the effective antitumor and radiosensitization effects via thermally aided chemodynamic/radiotreatment with a low radiation dose (6 Gy). Therefore, this work provides a potential strategy for overcoming the low-dose radioresistance in cancer therapy.
Collapse
Affiliation(s)
- Wensheng Xie
- The Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Jielin Ye
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Zhenhu Guo
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, P. R. China
- State Key Laboratory of Powder Metallurgy, Powder Metallurgy Research Institute, Central South University, Changsha 410083, P. R. China
| | - Jingsong Lu
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Xiaohan Gao
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, P. R. China
| | - Yen Wei
- The Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Lingyun Zhao
- State Key Laboratory of New Ceramics and Fine Processing, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, P. R. China
| |
Collapse
|
27
|
Cheng L, Jin X, Shen H, Chen X, Chen J, Xu B, Xu J. Icariin attenuates thioacetamide‑induced bone loss via the RANKL‑p38/ERK‑NFAT signaling pathway. Mol Med Rep 2022; 25:126. [PMID: 35169865 PMCID: PMC8864607 DOI: 10.3892/mmr.2022.12642] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 01/25/2022] [Indexed: 11/18/2022] Open
Abstract
There is an increasing incidence of destructive bone disease caused by osteoclast proliferation. This is characterized by reduced bone mass and imbalance of bone homeostasis. Icariin (ICA), a flavonoid compound isolated from Epimedium, has anti‑osteoporosis activity and inhibits the formation of osteoclasts and bone resorption. The purpose of the present study was to investigate the protective effect of ICA on osteoclastic differentiation induced by thioacetamide (TAA) and its possible mechanism in Sprague Dawley (SD) rats. In the present study, SD rats were intraperitoneally injected with TAA (300 mg/kg) for the bone loss model, treated with ICA (600 mg/kg, intragastric gavage) in the ICA group and TAA+ICA group for treatment of bone loss for 6 weeks. Indexes associated with bone metabolism, such as alkaline phosphatase, N‑terminal telopeptide of type‑I collagen (NTX‑I), calcium (Ca), phosphorus (P) and magnesium (Mg) in the serum, were detected. Osteoclast differentiation of femoral tissues was detected by hematoxylin and eosin and tartrate‑resistant acid phosphatase staining. The femoral bone mass was evaluated using a three‑point bending test and micro computed tomography. Western blotting was used to detect the expression levels of osteoclast‑related proteins in each group. In the rats treated with TAA, the serum concentrations of Ca, P and Mg were decreased, the serum concentration of NTX‑I was increased, osteoclast differentiation of the femur was increased, femur bone stress and bone mass were decreased and the bone loss and osteoclast formation were reduced after ICA treatment. In addition, ICA inhibited the protein expression of receptor activator of nuclear factor κ‑Β ligand (RANKL), receptor activator of nuclear factor κ‑B (RANK), p38, ERK, c‑Fos and nuclear factor of activated T cells 1 (NFATc1) in the femur of rats treated with TAA. The results suggested that ICA may inhibit osteoclast differentiation by downregulating the RANKL‑p38/ERK‑NFAT signaling pathway and prevent TAA‑induced bone loss. The results are helpful to understand the mechanism of osteoclast differentiation induced by TAA, as well as the antiresorptive activity and molecular mechanism of ICA, and to provide new ideas for the treatment of osteolytic diseases.
Collapse
Affiliation(s)
- Linyan Cheng
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Xiaoli Jin
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Hao Shen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Xuanwei Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Jin Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Bin Xu
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Jian Xu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| |
Collapse
|
28
|
Patwardhan RS, Sharma D, Sandur SK. Thioredoxin reductase: An emerging pharmacologic target for radiosensitization of cancer. Transl Oncol 2022; 17:101341. [PMID: 35078017 PMCID: PMC8790659 DOI: 10.1016/j.tranon.2022.101341] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 12/15/2022] Open
Abstract
Novel agents are required to increase the radiosensitivity of cancer and improve the outcome of radiotherapy. Thioredoxin (Trx) and thioredoxin reductase (TrxR) reduce the oxidized cysteine thiols in several proteins, which regulate cellular redox, survival, proliferation, DNA synthesis, transcription factor activity and apoptosis. TrxR is essential for maintaining a conducive redox state for tumor growth, survival and resistance to therapy. Therefore, it is an appealing pharmacological target for the radiosensitization of tumors. Ionizing radiation (IR) is known to cause cytotoxicity through ROS, oxidative stress and DNA damage. Inhibition of thioredoxin system augments IR induced oxidative stress and potentiates cytotoxic effects. However, TrxR also regulates several critical cellular processes in normal cells. Here, we highlight the pre-clinical research and pharmacological studies to surmise possible utility of different TrxR inhibitors for radiosensitization. This review provides a succinct perspective on the role of TrxR inhibitors during the radiotherapy of cancer.
Collapse
Affiliation(s)
- Raghavendra S Patwardhan
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Deepak Sharma
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India.
| | - Santosh K Sandur
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India.
| |
Collapse
|
29
|
Synthesis, Anticancer Assessment, and Molecular Docking of Novel Chalcone-Thienopyrimidine Derivatives in HepG2 and MCF-7 Cell Lines. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2021:4759821. [PMID: 35003514 PMCID: PMC8728392 DOI: 10.1155/2021/4759821] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/06/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022]
Abstract
Heterocycles containing thienopyrimidine moieties have attracted attention due to their interesting biological and pharmacological activities. In this research article, we reported the synthesis of a series of new hybrid molecules through merging the structural features of chalcones and pyridothienopyrimidinones. Our results indicated that the synthesis of chalcone-thienopyrimidine derivatives from the corresponding thienopyrimidine and chalcones proceeded in a relatively short reaction time with good yields and high purity. Most of these novel compounds exhibited moderate to robust cytotoxicity against HepG2 and MCF-7 cancer cells similar to that of 5-fluorouracil (5-FU). The results indicated that IC50 of the two compounds (3b and 3g) showed more potent anticancer activities against HepG2 and MCF-7 than 5-FU. An MTT assay and flow cytometry showed that only 3b and 3g had anticancer activity and antiproliferative activities at the G1 phase against MCF-7 cells, while six compounds (3a-e and 3g) had cytotoxicity and cell cycle arrest at different phases against HepG2 cells. Their cytotoxicity was achieved through downregulation of Bcl-2 and upregulation of Bax, caspase-3, and caspase-9. Although all tested compounds increased oxidative stress via increment of MDA levels and decrement of glutathione reductase (GR) activities compared to control, the 3a, 3b, and 3g in HepG2 and 3b and 3g in MCF-7 achieved the target results. Moreover, there was a positive correlation between cytotoxic efficacy of the compound and apoptosis in both HepG2 (R 2 = 0.531; P = 0.001) and MCF-7 (R 2 = 0.219; P = 0.349) cell lines. The results of molecular docking analysis of 3a-g into the binding groove of Bcl-2 revealed relatively moderate binding free energies compared to the selective Bcl-2 inhibitor, DRO. Like venetoclax, compounds 3a-g showed 2 violations from Lipinski's rule. However, the results of the ADME study also revealed higher drug-likeness scores for compounds 3a-g than for venetoclax. In conclusion, the tested newly synthesized chalcone-pyridothienopyrimidinone derivatives showed promising antiproliferative and apoptotic effects. Mechanistically, the compounds increased ROS production with concomitant cell cycle arrest and apoptosis. Therefore, regulation of the cell cycle and apoptosis are possible targets for anticancer therapy. The tested compounds could be potent anticancer agents to be tested in future clinical trials after extensive pharmacodynamic, pharmacokinetic, and toxicity profile investigations.
Collapse
|
30
|
Felicetti F, Aimaretti E, Dal Bello F, Gatti F, Godono A, Saba F, Einaudi G, Collino M, Fagioli F, Aragno M, Brignardello E. Advanced glycation end products and their related signaling cascades in adult survivors of childhood Hodgkin lymphoma: A possible role in the onset of late complications. Free Radic Biol Med 2022; 178:76-82. [PMID: 34856327 DOI: 10.1016/j.freeradbiomed.2021.11.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/23/2021] [Accepted: 11/28/2021] [Indexed: 12/12/2022]
Abstract
Hodgkin lymphoma (HL) is today one of the most curable pediatric cancers. Despite survival rates now exceeding 90%, survivors of pediatric HL are still at higher risk to develop late effects of cancer therapy. Premature aging has been proposed as a paradigm to explain the onset of long-term complications in these subjects. High levels of advanced glycation end products (AGEs), together with chronic inflammation and oxidative unbalance, have been shown to be among the main factors contributing to aging. The present study aims to evaluate glycoxydation, inflammatory status, and oxidative stress in plasma and peripheral blood mononuclear cells (PBMC) obtained from 20 adult survivors of pediatric HL and 40 age- and sex-matched healthy controls. After the isolation of PBMC and the collection of plasma, we performed the analyses of gene expression by qRT-PCR and measured inflammatory and oxidative-stress markers. AGEs plasma levels, expressed as Nϵ-carboxymethyl-lysine and methylglyoxal hydroimidazolone, were markedly higher in HL survivors than in healthy subjects. HL survivors also showed a condition of higher oxidative stress, as demonstrated by an increased expression of NADPH oxidase on PBMC. Antioxidant defenses, evaluated in terms of alpha-tocopherol, GSSG/GSH ratio and catalase plasma levels, were strongly impaired in survivors. This pro-oxidative condition led to the over-expression of both NLRP3 and NFkB genes in PBMC and, consequently, to increased plasma levels of interleukin(IL)-1β and IL-6. Finally, the expression of the receptors for AGEs in PBMC confirmed the dysregulated AGE pathways. Data show AGEs accumulation in survivors of pediatric HL. The consequent activation of the receptor for AGEs leads to the persistent activation of intracellular signaling toward inflammation. These results suggest that the co-existence of AGEs accumulation, unbalanced oxidative status, and inflammation could play a role in the onset of late complications in HL survivors.
Collapse
Affiliation(s)
- Francesco Felicetti
- Transition Unit for Childhood Cancer Survivors, Città della Salute e della Scienza Hospital, Turin, Italy
| | - Eleonora Aimaretti
- General Pathology Unit, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Federica Dal Bello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Italy
| | - Filippo Gatti
- Transition Unit for Childhood Cancer Survivors, Città della Salute e della Scienza Hospital, Turin, Italy
| | - Alessandro Godono
- Department of Public Health and Pediatrics, University of Torino, Turin, Italy
| | - Francesca Saba
- Department of Medical Science, University of Turin, Turin, Italy
| | - Giacomo Einaudi
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Massimo Collino
- Department of Neurosciences "Rita Levi Montalcini" University of Turin, Turin, Italy
| | - Franca Fagioli
- Department of Public Health and Pediatrics, University of Torino, Turin, Italy; Division of Paediatric Onco-Haematology, Stem Cell Transplantation and Cellular Therapy, Città della Salute e della Scienza Hospital, Turin, Italy
| | - Manuela Aragno
- General Pathology Unit, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Enrico Brignardello
- Transition Unit for Childhood Cancer Survivors, Città della Salute e della Scienza Hospital, Turin, Italy.
| |
Collapse
|
31
|
Jit BP, Pradhan B, Dash R, Bhuyan PP, Behera C, Behera RK, Sharma A, Alcaraz M, Jena M. Phytochemicals: Potential Therapeutic Modulators of Radiation Induced Signaling Pathways. Antioxidants (Basel) 2021; 11:antiox11010049. [PMID: 35052553 PMCID: PMC8773162 DOI: 10.3390/antiox11010049] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 02/06/2023] Open
Abstract
Ionizing radiation results in extensive damage to biological systems. The massive amount of ionizing radiation from nuclear accidents, radiation therapy (RT), space exploration, and the nuclear battlefield leads to damage to biological systems. Radiation injuries, such as inflammation, fibrosis, and atrophy, are characterized by genomic instability, apoptosis, necrosis, and oncogenic transformation, mediated by the activation or inhibition of specific signaling pathways. Exposure of tumors or normal cells to different doses of ionizing radiation could lead to the generation of free radical species, which can release signal mediators and lead to harmful effects. Although previous FDA-approved agents effectively mitigate radiation-associated toxicities, their use is limited due to their high cellular toxicities. Preclinical and clinical findings reveal that phytochemicals derived from plants that exhibit potent antioxidant activities efficiently target several signaling pathways. This review examined the prospective roles played by some phytochemicals in altering signal pathways associated with radiation response.
Collapse
Affiliation(s)
- Bimal Prasad Jit
- School of Life Sciences, Sambalpur University, Jyoti Vihar, Burla 768019, India; (B.P.J.); (R.D.); (R.K.B.)
- Department of Biochemistry, AIIMS, Ansari Nagar, New Delhi 110029, India;
| | - Biswajita Pradhan
- Algal Biotechnology and Molecular Systematic Laboratory, Post Graduate Department of Botany, Berhampur University, Bhanja Bihar, Berhampur 760007, India; (B.P.); (C.B.)
- Department of Biotechnology, Sangmyung University, Seoul 03016, Korea
| | - Rutumbara Dash
- School of Life Sciences, Sambalpur University, Jyoti Vihar, Burla 768019, India; (B.P.J.); (R.D.); (R.K.B.)
| | - Prajna Paramita Bhuyan
- Department of Botany, Maharaja Sriram Chandra Bhanja Deo University, Baripada 757003, India;
| | - Chhandashree Behera
- Algal Biotechnology and Molecular Systematic Laboratory, Post Graduate Department of Botany, Berhampur University, Bhanja Bihar, Berhampur 760007, India; (B.P.); (C.B.)
| | - Rajendra Kumar Behera
- School of Life Sciences, Sambalpur University, Jyoti Vihar, Burla 768019, India; (B.P.J.); (R.D.); (R.K.B.)
| | - Ashok Sharma
- Department of Biochemistry, AIIMS, Ansari Nagar, New Delhi 110029, India;
| | - Miguel Alcaraz
- Radiology and Physical Medicine Department, School of Medicine, Campus de Excelencia Internacional de Ámbito Regional (CEIR)-Campus Mare Nostrum (CMN), Universidad de Murcia, 30100 Murcia, Spain
- Correspondence: (M.A.); (M.J.); Tel.: +34-868883601 (M.A.); +91-7978478950 (M.J.)
| | - Mrutyunjay Jena
- Algal Biotechnology and Molecular Systematic Laboratory, Post Graduate Department of Botany, Berhampur University, Bhanja Bihar, Berhampur 760007, India; (B.P.); (C.B.)
- Correspondence: (M.A.); (M.J.); Tel.: +34-868883601 (M.A.); +91-7978478950 (M.J.)
| |
Collapse
|
32
|
Yang R, Tan C, Najafi M. Cardiac inflammation and fibrosis following chemo/radiation therapy: mechanisms and therapeutic agents. Inflammopharmacology 2021; 30:73-89. [PMID: 34813027 DOI: 10.1007/s10787-021-00894-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/08/2021] [Indexed: 12/14/2022]
Abstract
The incidence of cardiovascular disorders is one of the most concerns among people who underwent cancer therapy. The heart side effects of cancer therapy may occur during treatment to some years after the end of treatment. Some epidemiological studies confirm that heart diseases are one of the most common reasons for mortality among patients that were received treatment for cancer. Experimental studies and also clinical investigations indicate that inflammatory changes such as pericarditis, myocarditis, and also fibrosis are key mechanisms of cardiac diseases following chemotherapy/radiotherapy. It seems that chronic oxidative stress, massive cell death, and chronic overproduction of pro-inflammatory and pro-fibrosis cytokines are the key mechanisms of cardiovascular diseases following cancer therapy. Furthermore, infiltration of inflammatory cells and upregulation of some enzymes such as NADPH Oxidases are a hallmark of heart diseases after cancer therapy. In the current review, we aim to explain how radiation or chemotherapy can induce inflammatory and fibrosis-related diseases in the heart. We will explain the cellular and molecular mechanisms of cardiac inflammation and fibrosis following chemo/radiation therapy, and then review some adjuvants to reduce the risk of inflammation and fibrosis in the heart.
Collapse
Affiliation(s)
- Run Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, No. 139 Renmin Road, Changsha, Hunan, People's Republic of China
| | - Changming Tan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, No. 139 Renmin Road, Changsha, Hunan, People's Republic of China.
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
33
|
Patient-Derived Human Basal and Cutaneous Squamous Cell Carcinoma Tissues Display Apoptosis and Immunomodulation following Gas Plasma Exposure with a Certified Argon Jet. Int J Mol Sci 2021; 22:ijms222111446. [PMID: 34768877 PMCID: PMC8584092 DOI: 10.3390/ijms222111446] [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: 09/04/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 12/17/2022] Open
Abstract
Reactive oxygen species (ROS) have been subject of increasing interest in the pathophysiology and therapy of cancers in recent years. In skin cancer, ROS are involved in UV-induced tumorigenesis and its targeted treatment via, e.g., photodynamic therapy. Another recent technology for topical ROS generation is cold physical plasma, a partially ionized gas expelling dozens of reactive species onto its treatment target. Gas plasma technology is accredited for its wound-healing abilities in Europe, and current clinical evidence suggests that it may have beneficial effects against actinic keratosis. Since the concept of hormesis dictates that low ROS levels perform signaling functions, while high ROS levels cause damage, we investigated herein the antitumor activity of gas plasma in non-melanoma skin cancer. In vitro, gas plasma exposure diminished the metabolic activity, preferentially in squamous cell carcinoma cell (SCC) lines compared to non-malignant HaCaT cells. In patient-derived basal cell carcinoma (BCC) and SCC samples treated with gas plasma ex vivo, increased apoptosis was found in both cancer types. Moreover, the immunomodulatory actions of gas plasma treatment were found affecting, e.g., the expression of CD86 and the number of regulatory T-cells. The supernatants of these ex vivo cultured tumors were quantitatively screened for cytokines, chemokines, and growth factors, identifying CCL5 and GM-CSF, molecules associated with skin cancer metastasis, to be markedly decreased. These findings suggest gas plasma treatment to be an interesting future technology for non-melanoma skin cancer topical therapy.
Collapse
|
34
|
Li X, Wang Q, Yu S, Zhang M, Liu X, Deng G, Liu Y, Wu S. Multifunctional MnO 2-based nanoplatform-induced ferroptosis and apoptosis for synergetic chemoradiotherapy. Nanomedicine (Lond) 2021; 16:2343-2361. [PMID: 34523352 DOI: 10.2217/nnm-2021-0286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Background: Radiosensitizers that can effectively consume glutathione provide broad prospects for enhancing the efficacy and reducing the side effects of radiotherapy. Aim: To explore the potential role of CuS@mSiO2@MnO2 nanocomposites in synergetic chemoradiotherapy. Methods: Nanocomposites were characterized by transmission electron microscopy, UV-Vis spectrometry and dynamic light scattering and were loaded with doxorubicin (DOX). The uptake and biodistribution of nanocomposites were observed by CCK8 assay, MRI and confocal laser scanning microscopy. The radiosensitization effect of nanocomposites and nanocomposites/DOX was assessed both in vitro and in vivo. Results: In vitro application of nanocomposites, with an average diameter of 30 nm and ζ-potential of 13.2 ± 0.4 mV, in combination with radiotherapy, depleted glutathione and induced ferroptosis and apoptosis. Nanocomposites/DOX exhibited tumor cell damage in vivo. Conclusion: We propose that this glutathione-depleting nanosystem could be a radiosensitizer as well as a drug transporter.
Collapse
Affiliation(s)
- Xi Li
- Department of Obstetrics & Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 650 Xin Songjiang Road, Shanghai, 201620, China
| | - Qi Wang
- Department of Orthopedics, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200333, China.,Trauma Center, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 650 Xin Songjiang Road, Shanghai, 201620, China
| | - Sihui Yu
- Department of Obstetrics & Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 650 Xin Songjiang Road, Shanghai, 201620, China
| | - Minyi Zhang
- College of Chemistry & Chemical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - Xijian Liu
- College of Chemistry & Chemical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - Guoying Deng
- Trauma Center, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 650 Xin Songjiang Road, Shanghai, 201620, China
| | - Yuan Liu
- Reproductive Medicine Center, Department of Obstetrics & Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 650 Xin Songjiang Road, Shanghai, 201620, China
| | - Sufang Wu
- Department of Obstetrics & Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 650 Xin Songjiang Road, Shanghai, 201620, China
| |
Collapse
|
35
|
Perez-Gelvez YNC, Camus AC, Bridger R, Wells L, Rhodes OE, Bergmann CW. Effects of chronic exposure to low levels of IR on Medaka ( Oryzias latipes): a proteomic and bioinformatic approach. Int J Radiat Biol 2021; 97:1485-1501. [PMID: 34355643 DOI: 10.1080/09553002.2021.1962570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Chronic exposure to ionizing radiation (IR) at low doses (<100 mGy) has been insufficiently studied to understand fully the risk to health. Relatively little knowledge exists regarding how species and healthy tissues respond at the protein level to chronic exposure to low doses of IR, and mass spectrometric-based profiling of protein expression is a powerful tool for studying changes in protein abundance. MATERIALS AND METHODS SDS gel electrophoresis, LC-MS/MS mass spectrometry-based approaches and bioinformatic data analytics were used to detect proteomic changes following chronic exposure to moderate/low doses of radiation in adults and normally developed Medaka fish (Oryzias latipes). RESULTS Significant variations in the abundance of proteins involved in thyroid hormone signaling and lipid metabolism were detected, which could be related to the gonadal regression phenotype observed after 21.04 mGy and 204.3 mGy/day exposure. The global proteomic change was towards overexpression of proteins in muscle and skin, while the opposite effect was observed in internal organs. CONCLUSION The present study provides information on the impacts of biologically relevant low doses of IR, which will be useful in future research for the identification of potential biomarkers of IR exposure and allow for a better assessment of radiation biosafety regulations.
Collapse
Affiliation(s)
- Yeni Natalia C Perez-Gelvez
- Carbohydrate Complex Research Center, Biochemistry and Molecular Biology, The University of Georgia, Athens, GA, USA
| | - Alvin C Camus
- College of Veterinary Medicine, Department of Pathology, The University of Georgia, Athens, GA, USA
| | - Robert Bridger
- Carbohydrate Complex Research Center, The University of Georgia, Athens, GA, USA
| | - Lance Wells
- Carbohydrate Complex Research Center, The University of Georgia, Athens, GA, USA
| | - Olin E Rhodes
- Savannah River Ecology Laboratory, Odum School of Ecology, The University of Georgia, Athens, GA, USA
| | - Carl W Bergmann
- Carbohydrate Complex Research Center, The University of Georgia, Athens, GA, USA
| |
Collapse
|
36
|
Tumor Tissue Oxidative Stress Changes and Na, K-ATPase Evaluation in Head and Neck Squamous Cell Carcinoma. J Membr Biol 2021; 254:475-486. [PMID: 34104985 DOI: 10.1007/s00232-021-00185-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/08/2021] [Indexed: 10/21/2022]
Abstract
Changes in metabolism are mechanisms that are largely implicated in the development, progression, and metastasis of head and neck squamous cell carcinoma (HNSCC) and also in resistance to different anticancer therapies. Identification of biomarkers for differentiation between cancerous and normal epithelium, treatment design and prognosis remain a vital issue in the field of head and neck cancer. The present study analyzed the main biochemical changes that occur in HNSCC tumors by through mechanisms involving oxidative stress. The release of substances reactive to thiobarbituric acid was significantly lower in HNSCC tumor tissue as compared to healthy tissue. The assays related to the lipid profile assays showed changes in membrane biophysics of tumor cells due to an increase in total phospholipids and total cholesterol, as well as an increased activity and expression of the α1 subunit of Na, K-ATPase, which is fundamental in the process of carcinogenesis. The modulation of the antioxidant system was also affected, with a decrease in the catalytic activity of the enzymes superoxide dismutase (SOD) and glutathione peroxidase (GPx), as well as a reduction of glutathione (GSH) content and an increase in H2O2 content. A reduction in catalase (CAT) activity was observed. The data presented here are in accordance with important findings described by us in a previous study, involving the same individuals, but with a focus on the damage generated in red blood cells, resulting from tumor installation. Therefore, it was possible to conclude that the biochemical alterations found in HNSCC cells are fundamental for transformation and maintenance of the tumor cell and once it is installed, it is also capable of generating injuries in the patients' red blood cells. Our data demonstrate that this could be a promising biomarker for HNSCC.
Collapse
|
37
|
Szabo I, Zoratti M, Biasutto L. Targeting mitochondrial ion channels for cancer therapy. Redox Biol 2021; 42:101846. [PMID: 33419703 PMCID: PMC8113036 DOI: 10.1016/j.redox.2020.101846] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/19/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022] Open
Abstract
Pharmacological targeting of mitochondrial ion channels is emerging as a promising approach to eliminate cancer cells; as most of these channels are differentially expressed and/or regulated in cancer cells in comparison to healthy ones, this strategy may selectively eliminate the former. Perturbation of ion fluxes across the outer and inner membranes is linked to alterations of redox state, membrane potential and bioenergetic efficiency. This leads to indirect modulation of oxidative phosphorylation, which is/may be fundamental for both cancer and cancer stem cell survival. Furthermore, given the crucial contribution of mitochondria to intrinsic apoptosis, modulation of their ion channels leading to cytochrome c release may be of great advantage in case of resistance to drugs triggering apoptotic events upstream of the mitochondrial phase. In the present review, we give an overview of the known mitochondrial ion channels and of their modulators capable of killing cancer cells. In addition, we discuss state-of-the-art strategies using mitochondriotropic drugs or peptide-based approaches allowing a more efficient and selective targeting of mitochondrial ion channel-linked events.
Collapse
Affiliation(s)
- Ildiko Szabo
- Department of Biology, University of Padova, Italy; CNR Institute of Neurosciences, Padova, Italy.
| | | | - Lucia Biasutto
- CNR Institute of Neurosciences, Padova, Italy; Department of Biomedical Sciences, University of Padova, Italy
| |
Collapse
|
38
|
Farzaneh Z, Vosough M, Agarwal T, Farzaneh M. Critical signaling pathways governing hepatocellular carcinoma behavior; small molecule-based approaches. Cancer Cell Int 2021; 21:208. [PMID: 33849569 PMCID: PMC8045321 DOI: 10.1186/s12935-021-01924-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of death due to cancer. Although there are different treatment options, these strategies are not efficient in terms of restricting the tumor cell's proliferation and metastasis. The liver tumor microenvironment contains the non-parenchymal cells with supportive or inhibitory effects on the cancerous phenotype of HCC. Several signaling pathways are dis-regulated in HCC and cause uncontrolled cell propagation, metastasis, and recurrence of liver carcinoma cells. Recent studies have established new approaches for the prevention and treatment of HCC using small molecules. Small molecules are compounds with a low molecular weight that usually inhibit the specific targets in signal transduction pathways. These components can induce cell cycle arrest, apoptosis, block metastasis, and tumor growth. Devising strategies for simultaneously targeting HCC and the non-parenchymal population of the tumor could lead to more relevant research outcomes. These strategies may open new avenues for the treatment of HCC with minimal cytotoxic effects on healthy cells. This study provides the latest findings on critical signaling pathways governing HCC behavior and using small molecules in the control of HCC both in vitro and in vivo models.
Collapse
Affiliation(s)
- Zahra Farzaneh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
39
|
HNC0014, a Multi-Targeted Small-Molecule, Inhibits Head and Neck Squamous Cell Carcinoma by Suppressing c-Met/STAT3/CD44/PD-L1 Oncoimmune Signature and Eliciting Antitumor Immune Responses. Cancers (Basel) 2020; 12:cancers12123759. [PMID: 33327484 PMCID: PMC7764918 DOI: 10.3390/cancers12123759] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Cancer stem cells (CSCs) in head and neck squamous cell carcinoma (HNSCC) possess unlimited self-renewal capacity, resist treatments and induce tumor repopulation after interventions. Here, we observed HNSCC CSCs secreted exosomes containing c-Met, STAT3 (also the phosphorylated form of c-Met and STAT3), CD44, and PD-L1 oncogenic signaling molecules. CSC-derived exosomes, in part, transform fibroblasts (NFs) into cancer-associated fibroblasts (CAFs), establish drug resistance, and an immune-evasive tumor microenvironment (TME). We demonstrated HNC0014, a novel small-molecule drug, suppresses HNSCC tumorigenesis, CSC generation and prevents CAF transformation by decreasing the aforementioned oncogenic signaling molecules’ expression in both HNSCC cells and CSC-derived exosomes. Abstract Despite advancements in diagnostic and standard treatment modalities, including surgery, radiotherapy, and chemotherapy, overall survival rates of advanced-stage head and neck squamous cell carcinoma (HNSCC) patients have remained stagnant for over three decades. Failure of these treatment modalities, coupled with post-therapy complications, underscores the need for alternative interventions and an in-depth understanding of the complex signaling networks involved in developing treatment resistance. Using bioinformatics tools, we identified an increased expression of c-Met, STAT3, and CD44 corresponding to a poor prognosis and malignant phenotype of HNSCC. Subsequently, we showed that tumorsphere-derived exosomes promoted cisplatin (CDDP) resistance and colony and tumorsphere formation in parental HNSCC cells, accompanied by an increased level of oncogenic/immune evasive markers, namely, c-Met, STAT3, CD44, and PD-L1. We then evaluated the therapeutic potential of a new small molecule, HNC0014. The molecular docking analysis suggested strong interactions between HNC0014 and oncogenic molecules; c-Met, STAT3, CD44, and PD-L1. Subsequently, we demonstrated that HNC0014 treatment suppressed HNSCC tumorigenic and expression of stemness markers; HNC0014 also reduced cancer-associated fibroblast (CAF) transformation by Exosp- and CAF-induced tumorigenic properties. HNC0014 treatment alone suppressed tumor growth in a cisplatin-resistant (SAS tumorspheres) mouse xenograft model and with higher inhibitory efficacy when combined with CDDP. More importantly, HNC0014 treatment significantly delayed tumor growth in a syngeneic mouse HNSCC model, elicited an antitumor immune profile, and reduced the total c-Met, STAT3, and their phosphorylated forms, PD-L1 and CD44, contents in serum exosomes. Collectively, our findings provide supports for HNC0014 as a multi-targeted immunotherapeutic lead compound for further development.
Collapse
|
40
|
Chang HW, Lee M, Lee YS, Kim SH, Lee JC, Park JJ, Nam HY, Kim MR, Han MW, Kim SW, Kim SY. p53-dependent glutamine usage determines susceptibility to oxidative stress in radioresistant head and neck cancer cells. Cell Signal 2020; 77:109820. [PMID: 33137455 DOI: 10.1016/j.cellsig.2020.109820] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 01/08/2023]
Abstract
The manner in which p53 maintains redox homeostasis and the means by which two key metabolic elements, glucose and glutamine, contribute to p53-dependent redox stability remain unclear. To elucidate the manner in which p53 deals with glucose-deprived, reactive oxygen species (ROS)-prone conditions in this regard, two isogenic cancer subclones (HN3R-A and HN3R-B) bearing distinct p53 mutations as an in vitro model of intratumoral p53 heterogeneity were identified. Following cumulative irradiation, the subclones showed a similar metabolic shift to aerobic glycolysis and increasing NADPH biogenesis for cellular defense against oxidative damage irrespective of p53 status. The radioresistant cancer cells became more sensitive to glycolysis-targeting drugs. However, in glucose-deprived and ROS-prone conditions, HN3R-B, the subclone with the original p53 increased the utilization of glutamine by GLS2, thereby maintaining redox homeostasis and ATP. Conversely, HN3R-A, the p53-deficient radioresistant subclone displayed an impairment in glutamine usage and high susceptibility to metabolic stresses as well as ROS-inducing agents despite the increased ROS scavenging system. Collectively, our findings suggest that p53 governs the alternative utilization of metabolic ingredients, such as glucose and glutamine, in ROS-prone conditions. Thus, p53 status may be an important biomarker for selecting cancer treatment strategies, including metabolic drugs and ROS-inducing agents, for recurrent cancers after radiotherapy.
Collapse
Affiliation(s)
- Hyo Won Chang
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - MyungJin Lee
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yoon Sun Lee
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Song Hee Kim
- Department of Otolaryngology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Jong Cheol Lee
- Department of Otolaryngology, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, South Korea
| | - Jung Je Park
- Department of Otolaryngology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Hae Yun Nam
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Mi Ra Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Inje University College of Medicine, Haeundae Paik Hospital, Busan, Republic of Korea
| | - Myung Woul Han
- Department of Otolaryngology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Seong Who Kim
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Sang Yoon Kim
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|