1
|
Hiruma K, Bilim V, Kazama A, Shirono Y, Murata M, Tomita Y. Acidic Microenvironment Enhances Cisplatin Resistance in Bladder Cancer via Bcl-2 and XIAP. Curr Issues Mol Biol 2025; 47:43. [PMID: 39852158 PMCID: PMC11763506 DOI: 10.3390/cimb47010043] [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: 12/23/2024] [Revised: 01/04/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
Cisplatin (CDDP) remains a key drug for patients with advanced bladder cancer (BC), despite the emergence of new therapeutic agents; thus, the identification of factors contributing to CDDP treatment resistance is crucial. As acidity of the tumor microenvironment has been reported to be associated with treatment resistance and poor prognosis across various cancer types, our objectives in this study were to investigate the effects of an acidic environment on BC cells and elucidate the mechanisms behind CDDP resistance. Our findings show that BC cells cultured under acidic conditions developed cisplatin resistance as acidity increased. Notably, CDDP administered to BC cells in a pH 6.0 environment required double the concentration, compared to those in a pH 7.5 environment, to achieve equivalent toxicity. Using chloroquine and navitoclax, we identified the involvement of the Bcl-2 and LC3B pathways in the acquisition of CDDP resistance under acidic conditions. A Western blot analysis revealed that the activations of Bcl-2 and XIAP expression appear to inhibit both apoptotic and autophagic cell death. Taken together, these results suggest that alleviating the acidity of the tumor microenvironment in clinical settings might enhance BC sensitivity to CDDP.
Collapse
Affiliation(s)
- Kaede Hiruma
- Department of Urology, Division of Molecular Oncology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; (V.B.)
| | - Vladimir Bilim
- Department of Urology, Division of Molecular Oncology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; (V.B.)
- Department of Urology, Kameda Daiichi Hospital, Niigata 950-0165, Japan
| | - Akira Kazama
- Department of Urology, Division of Molecular Oncology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; (V.B.)
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Yuko Shirono
- Department of Urology, Division of Molecular Oncology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; (V.B.)
- Department of Urology, Niigata Cancer Center Hospital, Niigata 951-8133, Japan
| | - Masaki Murata
- Department of Urology, Division of Molecular Oncology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; (V.B.)
- Department of Urology, Niigata Prefectural Central Hospital, Niigata 943-0192, Japan
| | - Yoshihiko Tomita
- Department of Urology, Division of Molecular Oncology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan; (V.B.)
| |
Collapse
|
2
|
Pang X, Li J, Liu Z, Chen Q, Chen WH, Zhang XZ, Luo GF, Shang Z. Targeted Elimination of the Oral Pathogen to Overcome Chemoresistance of Oral Squamous Cell Carcinoma by Biologically Derived Nanotherapeutics. ACS NANO 2024; 18:31794-31808. [PMID: 39513690 DOI: 10.1021/acsnano.4c07000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Local oral microbiota are closely related to the tumorigenesis and therapeutic response of oral cancer. In this study, we have validated that oral commensal Porphyromonas gingivalis (P. gingivalis) is highly responsible for chemoresistance and contributes to the poor therapeutic outcome of traditional chemotherapy. Accordingly, the biologically derived nanovesicles from ginger (GDNVs) with excellent P. gingivalis elimination ability are explored to transport the clinically used drug paclitaxel (PTX) for potentiating the therapeutic efficiency. Taking advantage of active targeting and inhibition abilities of GDNVs against P. gingivalis, the PTX-loaded GDNVs nanosystem (P-GDNVs) can enrich in the P. gingivalis-colonized tumor tissues and effectively inhibit the growth of P. gingivalis for downregulating the IL-6/pSTAT3/P-gp pathway, thereby reducing the efflux of intracellular drugs to overcome chemoresistance. By evaluating both P. gingivalis-infected tumor cells and P. gingivalis-infiltrated tumor-bearing mice, P-GDNVs show a much enhanced tumor cell killing effect, as compared with free PTX. This naturally occurring nanotherapeutic system represents an effective bioactive material for targeted elimination of host microbiota to boost therapeutic response, showing great promise to combat commensal microbiota-rich tumors.
Collapse
MESH Headings
- Porphyromonas gingivalis/drug effects
- Mouth Neoplasms/drug therapy
- Mouth Neoplasms/microbiology
- Mouth Neoplasms/pathology
- Mouth Neoplasms/metabolism
- Animals
- Mice
- Paclitaxel/pharmacology
- Paclitaxel/chemistry
- Drug Resistance, Neoplasm/drug effects
- Humans
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/microbiology
- Cell Line, Tumor
- Mice, Inbred BALB C
- Nanoparticles/chemistry
- Cell Proliferation/drug effects
- Antineoplastic Agents, Phytogenic/pharmacology
- Antineoplastic Agents, Phytogenic/chemistry
- Mice, Nude
- Drug Screening Assays, Antitumor
Collapse
Affiliation(s)
- Xiaochan Pang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China
| | - Ji Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China
| | - Zhenan Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China
| | - Qingli Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China
| | - Wei-Hai Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Guo-Feng Luo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China
- Hubei Key Laboratory of Regenerative Medicine and Multi-disciplinary Translational Research, Wuhan 430022, China
| | - Zhengjun Shang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, P. R. China
| |
Collapse
|
3
|
Li D, Shao F, Yu Q, Wu R, Tuo Z, Wang J, Ye L, Guo Y, Yoo KH, Ke M, Okoli UA, Premkamon C, Yang Y, Wei W, Heavey S, Cho WC, Feng D. The complex interplay of tumor-infiltrating cells in driving therapeutic resistance pathways. Cell Commun Signal 2024; 22:405. [PMID: 39160622 PMCID: PMC11331645 DOI: 10.1186/s12964-024-01776-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/01/2024] [Indexed: 08/21/2024] Open
Abstract
Drug resistance remains a significant challenge in cancer treatment. Recently, the interactions among various cell types within the tumor microenvironment (TME) have deepened our understanding of the mechanisms behind treatment resistance. Therefore, this review aims to synthesize current research focusing on infiltrating cells and drug resistance suggesting that targeting the TME could be a viable strategy to combat this issue. Numerous factors, including inflammation, metabolism, senescence, hypoxia, and angiogenesis, contribute to drug resistance could be a viable strategy to combat this issue. Overexpression of STAT3 is commonly associated with drug-resistant cancer cells or stromal cells. Current research often generalizes the impact of stromal cells on resistance, lacking specificity and statistical robustness. Thus, future research should take notice of this issue and aim to provide high-quality evidence. Despite the existing limitations, targeting the TME to overcome therapy resistance hold promising and valuable potential.
Collapse
Affiliation(s)
- Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fanglin Shao
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Qingxin Yu
- Department of Pathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo, 315211, China
- Department of Pathology, Ningbo Medical Centre Lihuili Hospital, Ningbo, China
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhouting Tuo
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Luxia Ye
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Yiqing Guo
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, Seoul, Republic of Korea
| | - Mang Ke
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
- Department of Urology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Uzoamaka Adaobi Okoli
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK
- Basic and Translational Cancer Research Group, Department of Pharmacology and Therapeutics, College of Medicine, University of Nigeria, Eastern part of Nigeria, Nsukka, Enugu State, Nigeria
| | - Chaipanichkul Premkamon
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK
| | - Yubo Yang
- Department of Urology, Three Gorges Hospital, Chongqing University, Wanzhou, Chongqing, 404000, China
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Susan Heavey
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK.
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Birmingham, Hong Kong SAR, China.
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK.
| |
Collapse
|
4
|
Lagzian A, Askari M, Haeri MS, Sheikhi N, Banihashemi S, Nabi-Afjadi M, Malekzadegan Y. Increased V-ATPase activity can lead to chemo-resistance in oral squamous cell carcinoma via autophagy induction: new insights. Med Oncol 2024; 41:108. [PMID: 38592406 DOI: 10.1007/s12032-024-02313-9] [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: 11/27/2023] [Accepted: 01/23/2024] [Indexed: 04/10/2024]
Abstract
Oral squamous cell carcinoma (OSCC) is a cancer type with a high rate of recurrence and a poor prognosis. Tumor chemo-resistance remains an issue for OSCC patients despite the availability of multimodal therapy options, which causes an increase in tumor invasiveness. Vacuolar ATPase (V-ATPase), appears to be one of the most significant molecules implicated in MDR in tumors like OSCC. It is primarily responsible for controlling the acidity in the solid tumors' microenvironment, which interferes with the absorption of chemotherapeutic medications. However, the exact cellular and molecular mechanisms V-ATPase plays in OSCC chemo-resistance have not been understood. Uncovering these mechanisms can contribute to combating OSCC chemo-resistance and poor prognosis. Hence, in this review, we suggest that one of these underlying mechanisms is autophagy induced by V-ATPase which can potentially contribute to OSCC chemo-resistance. Finally, specialized autophagy and V-ATPase inhibitors may be beneficial as an approach to reduce drug resistance to anticancer therapies in addition to serving as coadjuvants in antitumor treatments. Also, V-ATPase could be a prognostic factor for OSCC patients. However, in the future, more investigations are required to demonstrate these suggestions and hypotheses.
Collapse
Affiliation(s)
- Ahmadreza Lagzian
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Marziye Askari
- Department of Immunology, School of Medicine, Hamedan University of Medical Sciences, Hamedan, Iran
| | - Melika Sadat Haeri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Nastaran Sheikhi
- Biotechnology Department, Biological Sciences Faculty, Alzahra University, Tehran, Iran
| | - Sara Banihashemi
- Department of Bioscience, School of Science and Technology, Nottingham Trend University, Nottingham, UK
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Yalda Malekzadegan
- Department of Microbiology, Saveh University of Medical Sciences, Saveh, Iran.
| |
Collapse
|
5
|
Shie WY, Cheng SJ, Chen KC, Tang CC, Peng HH, Ko HH, Hou HH, Elizabeth Chou HY. Fibroblast growth factor 5 expression predicts the progression of oral squamous cell carcinoma. J Formos Med Assoc 2024; 123:390-399. [PMID: 37704482 DOI: 10.1016/j.jfma.2023.08.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/05/2023] [Accepted: 08/15/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND/PURPOSE Fibroblast growth factor (FGF) 5 is a member of the FGF family that functions as a regulator of tissue growth and regeneration. Aberrant FGF5 expression has been previously associated with the progression of a number of different malignancies. However, its potential role in oral cancer remains unclear. In this study, we explored the relationship between the expression of FGF5 protein in oral squamous cell carcinomas (OSCCs) and the clinicopathological parameters of OSCCs and whether the expression of FGF5 protein in OSCCs could be a prognostic factor for OSCC patients. METHODS The FGF5 protein expression was examined in 64 OSCC and 34 normal oral mucosal specimens by immunohistochemical staining. Stress induced upregulation and intracellular redistribution of FGF5 were verified using xenograft animal model and OSCC cell lines. RESULTS The mean FGF5 protein labelling index was significantly higher in OSCC than in normal oral mucosal samples, with high FGF5 protein labelling index (>58%) being correlated with advanced stage and poor survival of OSCC patients. Apart from the peri-cytoplasmic staining pattern characteristic of paracrine growth factors, FGF5 protein was localized as distinct punctate structures in the cytoplasm of advanced stage or stressed-induced cells. This redistribution and upregulation of FGF5 protein could be sustained after termination of the stress induction in cell line and xenograft animal models. CONCLUSION FGF5 can be induced by cellular stress and risk factors of OSCC, where high expression levels of FGF5 is potentially a useful parameter for predicting OSCC progression and patient survival.
Collapse
Affiliation(s)
- Wan-Yi Shie
- Graduate Institute of Oral Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shih-Jung Cheng
- Graduate Institute of Oral Biology, College of Medicine, National Taiwan University, Taipei, Taiwan; School of Dentistry, National Taiwan University, Taipei, Taiwan; Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan; Department of Dentistry, National Taiwan University Hospital, College of Medicine, Taipei, Taiwan
| | - Kuan-Chi Chen
- Graduate Institute of Oral Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ching-Chun Tang
- Graduate Institute of Oral Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Hui Peng
- School of Dentistry, National Taiwan University, Taipei, Taiwan; Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan; Department of Dentistry, National Taiwan University Hospital, College of Medicine, Taipei, Taiwan; Department of Dentistry, National Taiwan University Hospital Hsin-Chu Branch, College of Medicine, Hsin-Chu, Taiwan
| | - Hui-Hsin Ko
- School of Dentistry, National Taiwan University, Taipei, Taiwan; Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan; Department of Dentistry, National Taiwan University Hospital, College of Medicine, Taipei, Taiwan; Department of Dentistry, National Taiwan University Hospital Hsin-Chu Branch, College of Medicine, Hsin-Chu, Taiwan
| | - Hsin-Han Hou
- Graduate Institute of Oral Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Han-Yi Elizabeth Chou
- Graduate Institute of Oral Biology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Dentistry, National Taiwan University Hospital, College of Medicine, Taipei, Taiwan; Center for Biotechnology, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
6
|
Mishra R. Oral tumor heterogeneity, its implications for patient monitoring and designing anti-cancer strategies. Pathol Res Pract 2024; 253:154953. [PMID: 38039738 DOI: 10.1016/j.prp.2023.154953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/11/2023] [Accepted: 11/15/2023] [Indexed: 12/03/2023]
Abstract
Oral cancer tumors occur in the mouth and are mainly derived from oral mucosa linings. It is one of the most common and fatal malignant diseases worldwide. The intratumor heterogeneity (ITH) of oral cancerous tumor is vast, so it is challenging to study and interpret. Due to environmental selection pressures, ITH arises through diverse genetic, epigenetic, and metabolic alterations. The ITH also talks about peri-tumoral vascular/ lymphatic growth, perineural permeation, tumor necrosis, invasion, and clonal expansion/ the coexistence of multiple subclones in a single tumor. The heterogeneity offers tumors the adaptability to survive, induce growth/ metastasis, and, most importantly, escape antitumor therapy. Unfortunately, the ITH is prioritized less in determining disease pathology than the traditional TNM classifications or tumor grade. Understanding ITH is challenging, but with the advancement of technology, this ITH can be decoded. Tumor genomics, proteomics, metabolomics, and other modern analyses can provide vast information. This information in clinics can assist in understanding a tumor's severity and be used for diagnostic, prognostic, and therapeutic decision-making. Lastly, the oral tumor ITH can lead to individualized, targeted therapy strategies fighting against OC.
Collapse
Affiliation(s)
- Rajakishore Mishra
- Department of Life Sciences, School of Natural Sciences, Central University of Jharkhand, Cheri-Manatu, Kamre, Ranchi 835 222, Jharkhand, India.
| |
Collapse
|
7
|
Davodabadi F, Sajjadi SF, Sarhadi M, Mirghasemi S, Nadali Hezaveh M, Khosravi S, Kamali Andani M, Cordani M, Basiri M, Ghavami S. Cancer chemotherapy resistance: Mechanisms and recent breakthrough in targeted drug delivery. Eur J Pharmacol 2023; 958:176013. [PMID: 37633322 DOI: 10.1016/j.ejphar.2023.176013] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 08/28/2023]
Abstract
Conventional chemotherapy, one of the most widely used cancer treatment methods, has serious side effects, and usually results in cancer treatment failure. Drug resistance is one of the primary reasons for this failure. The most significant drawbacks of systemic chemotherapy are rapid clearance from the circulation, the drug's low concentration in the tumor site, and considerable adverse effects outside the tumor. Several ways have been developed to boost neoplasm treatment efficacy and overcome medication resistance. In recent years, targeted drug delivery has become an essential therapeutic application. As more mechanisms of tumor treatment resistance are discovered, nanoparticles (NPs) are designed to target these pathways. Therefore, understanding the limitations and challenges of this technology is critical for nanocarrier evaluation. Nano-drugs have been increasingly employed in medicine, incorporating therapeutic applications for more precise and effective tumor diagnosis, therapy, and targeting. Many benefits of NP-based drug delivery systems in cancer treatment have been proven, including good pharmacokinetics, tumor cell-specific targeting, decreased side effects, and lessened drug resistance. As more mechanisms of tumor treatment resistance are discovered, NPs are designed to target these pathways. At the moment, this innovative technology has the potential to bring fresh insights into cancer therapy. Therefore, understanding the limitations and challenges of this technology is critical for nanocarrier evaluation.
Collapse
Affiliation(s)
- Fatemeh Davodabadi
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran.
| | - Seyedeh Fatemeh Sajjadi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran.
| | - Mohammad Sarhadi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Shaghayegh Mirghasemi
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Mahdieh Nadali Hezaveh
- Department of Chemical Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran.
| | - Samin Khosravi
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Mahdieh Kamali Andani
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran.
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain.
| | - Mohsen Basiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Saeid Ghavami
- Academy of Silesia, Faculty of Medicine, Rolna 43, 40-555. Katowice, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 3P5, Canada.
| |
Collapse
|
8
|
Dorna D, Paluszczak J. Targeting cancer stem cells as a strategy for reducing chemotherapy resistance in head and neck cancers. J Cancer Res Clin Oncol 2023; 149:13417-13435. [PMID: 37453969 PMCID: PMC10587253 DOI: 10.1007/s00432-023-05136-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
PURPOSE Resistance to chemotherapy and radiotherapy is the primary cause of a poor prognosis in oncological patients. Researchers identified many possible mechanisms involved in gaining a therapy-resistant phenotype by cancer cells, including alterations in intracellular drug accumulation, detoxification, and enhanced DNA damage repair. All these features are characteristic of stem cells, making them the major culprit of chemoresistance. This paper reviews the most recent evidence regarding the association between the stemness phenotype and chemoresistance in head and neck cancers. It also investigates the impact of pharmacologically targeting cancer stem cell populations in this subset of malignancies. METHODS This narrative review was prepared based on the search of the PubMed database for relevant papers. RESULTS Head and neck cancer cells belonging to the stem cell population are distinguished by the high expression of certain surface proteins (e.g., CD10, CD44, CD133), pluripotency-related transcription factors (SOX2, OCT4, NANOG), and increased activity of aldehyde dehydrogenase (ALDH). Chemotherapy itself increases the percentage of stem-like cells. Importantly, the intratumor heterogeneity of stem cell subpopulations reflects cell plasticity which has great importance for chemoresistance induction. CONCLUSIONS Evidence points to the advantage of combining classical chemotherapeutics with stemness modulators thanks to the joint targeting of the bulk of proliferating tumor cells and chemoresistant cancer stem cells, which could cause recurrence.
Collapse
Affiliation(s)
- Dawid Dorna
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Ul. Święcickiego 4, 60-781 Poznan, Poland
| | - Jarosław Paluszczak
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Ul. Święcickiego 4, 60-781 Poznan, Poland
| |
Collapse
|
9
|
Siquara da Rocha LDO, Souza BSDF, Coletta RD, Lambert DW, Gurgel Rocha CA. Mapping Cell-in-Cell Structures in Oral Squamous Cell Carcinoma. Cells 2023; 12:2418. [PMID: 37830632 PMCID: PMC10572403 DOI: 10.3390/cells12192418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/10/2023] [Accepted: 09/11/2023] [Indexed: 10/14/2023] Open
Abstract
Cell-in-cell (CIC) structures contribute to tumor aggressiveness and poor prognosis in oral squamous cell carcinoma (OSCC). In vitro 3D models may contribute to the understanding of the underlying molecular mechanisms of these events. We employed a spheroid model to study the CIC structures in OSCC. Spheroids were obtained from OSCC (HSC3) and cancer-associated fibroblast (CAF) lines using the Nanoshuttle-PLTM bioprinting system (Greiner Bio-One). Spheroid form, size, and reproducibility were evaluated over time (EvosTM XL; ImageJ version 1.8). Slides were assembled, stained (hematoxylin and eosin), and scanned (Axio Imager Z2/VSLIDE) using the OlyVIA System (Olympus Life Science) and ImageJ software (NIH) for cellular morphology and tumor zone formation (hypoxia and/or proliferative zones) analysis. CIC occurrence, complexity, and morphology were assessed considering the spheroid regions. Well-formed spheroids were observed within 6 h of incubation, showing the morphological aspects of the tumor microenvironment, such as hypoxic (core) and proliferative zone (periphery) formation. CIC structures were found in both homotypic and heterotypic groups, predominantly in the proliferative zone of the mixed HSC3/CAF spheroids. "Complex cannibalism" events were also noted. These results showcase the potential of this model in further studies on CIC morphology, formation, and relationship with tumor prognosis.
Collapse
Affiliation(s)
- Leonardo de Oliveira Siquara da Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil; (L.d.O.S.d.R.); (B.S.d.F.S.)
- Department of Pathology and Forensic Medicine, School of Medicine, Federal University of Bahia, Salvador 40110-100, BA, Brazil
| | - Bruno Solano de Freitas Souza
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil; (L.d.O.S.d.R.); (B.S.d.F.S.)
- D’Or Institute for Research and Education (IDOR), Salvador 41253-190, BA, Brazil
| | - Ricardo Della Coletta
- Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba 13414-903, SP, Brazil
- Graduate Program in Oral Biology, School of Dentistry, University of Campinas, Piracicaba 13414-903, SP, Brazil
| | - Daniel W. Lambert
- School of Clinical Dentistry, The University of Sheffield, Sheffield S10 2TA, UK
| | - Clarissa A. Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, BA, Brazil; (L.d.O.S.d.R.); (B.S.d.F.S.)
- Department of Pathology and Forensic Medicine, School of Medicine, Federal University of Bahia, Salvador 40110-100, BA, Brazil
- D’Or Institute for Research and Education (IDOR), Salvador 41253-190, BA, Brazil
- Department of Propaedeutics, School of Dentistry, Federal University of Bahia, Salvador 40110-150, BA, Brazil
| |
Collapse
|
10
|
Yoon K, Jung S, Ryu J, Park HJ, Oh HK, Kook MS. Redox-Sensitive Delivery of Doxorubicin from Nanoparticles of Poly(ethylene glycol)-Chitosan Copolymer for Treatment of Drug-Resistant Oral Cancer Cells. Int J Mol Sci 2023; 24:13704. [PMID: 37762003 PMCID: PMC10531032 DOI: 10.3390/ijms241813704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
Reactive oxygen species (ROS)-sensitive polymer nanoparticles were synthesized for tumor targeting of an anticancer drug, doxorubicin (DOX). For this purpose, chitosan-methoxy poly(ethylene glycol) (mPEG) (ChitoPEG)-graft copolymer was synthesized and then DOX was conjugated to the backbone of chitosan using a thioketal linker. Subsequently, the chemical structure of the DOX-conjugated ChitoPEG copolymer (ChitoPEGthDOX) was confirmed via 1H nuclear magnetic resonance (NMR) spectra. Nanoparticles of the ChitoPEGthDOX conjugates have spherical shapes and a size of approximately 100 nm. Transmission electron microscopy (TEM) has shown that ChitoPEGthDOX nanoparticles disintegrate in the presence of hydrogen peroxide and the particle size distribution also changes from a monomodal/narrow distribution pattern to a multi-modal/wide distribution pattern. Furthermore, DOX is released faster in the presence of hydrogen peroxide. These results indicated that ChitoPEGthDOX nanoparticles have ROS sensitivity. The anticancer activity of the nanoparticles was evaluated using AT84 oral squamous carcinoma cells. Moreover, DOX-resistant AT84 cells were prepared in vitro. DOX and its nanoparticles showed dose-dependent cytotoxicity in both DOX-sensitive and DOX-resistant AT84 cells in vitro. However, DOX itself showed reduced cytotoxicity against DOX-resistant AT84 cells, while the nanoparticles showed almost similar cytotoxicity to DOX-sensitive and DOX-resistant AT84 cells. This result may be due to the inhibition of intracellular delivery of free DOX, while nanoparticles were efficiently internalized in DOX-resistant cells. The in vivo study of a DOX-resistant AT84 cell-bearing tumor xenograft model showed that nanoparticles have higher antitumor efficacy than those found in free DOX treatment. These results may be related to the efficient accumulation of nanoparticles in the tumor tissue, i.e., the fluorescence intensity in the tumor tissue was stronger than that of any other organs. Our findings suggest that ChitoPEGthDOX nanoparticles may be a promising candidate for ROS-sensitive anticancer delivery against DOX-resistant oral cancer cells.
Collapse
Affiliation(s)
- Kaengwon Yoon
- El-Dental Clinic, Seomun Daero Street 625, Namgu, Gwangju 61737, Republic of Korea;
- Department of Maxillofacial Oral Surgery, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea; (S.J.); (J.R.); (H.-J.P.); (H.-K.O.)
| | - Seunggon Jung
- Department of Maxillofacial Oral Surgery, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea; (S.J.); (J.R.); (H.-J.P.); (H.-K.O.)
| | - Jaeyoung Ryu
- Department of Maxillofacial Oral Surgery, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea; (S.J.); (J.R.); (H.-J.P.); (H.-K.O.)
| | - Hong-Ju Park
- Department of Maxillofacial Oral Surgery, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea; (S.J.); (J.R.); (H.-J.P.); (H.-K.O.)
| | - Hee-Kyun Oh
- Department of Maxillofacial Oral Surgery, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea; (S.J.); (J.R.); (H.-J.P.); (H.-K.O.)
| | - Min-Suk Kook
- Department of Maxillofacial Oral Surgery, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea; (S.J.); (J.R.); (H.-J.P.); (H.-K.O.)
| |
Collapse
|
11
|
Bou Antoun N, Chioni AM. Dysregulated Signalling Pathways Driving Anticancer Drug Resistance. Int J Mol Sci 2023; 24:12222. [PMID: 37569598 PMCID: PMC10418675 DOI: 10.3390/ijms241512222] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
One of the leading causes of death worldwide, in both men and women, is cancer. Despite the significant development in therapeutic strategies, the inevitable emergence of drug resistance limits the success and impedes the curative outcome. Intrinsic and acquired resistance are common mechanisms responsible for cancer relapse. Several factors crucially regulate tumourigenesis and resistance, including physical barriers, tumour microenvironment (TME), heterogeneity, genetic and epigenetic alterations, the immune system, tumour burden, growth kinetics and undruggable targets. Moreover, transforming growth factor-beta (TGF-β), Notch, epidermal growth factor receptor (EGFR), integrin-extracellular matrix (ECM), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), phosphoinositol-3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR), wingless-related integration site (Wnt/β-catenin), Janus kinase/signal transducers and activators of transcription (JAK/STAT) and RAS/RAF/mitogen-activated protein kinase (MAPK) signalling pathways are some of the key players that have a pivotal role in drug resistance mechanisms. To guide future cancer treatments and improve results, a deeper comprehension of drug resistance pathways is necessary. This review covers both intrinsic and acquired resistance and gives a comprehensive overview of recent research on mechanisms that enable cancer cells to bypass barriers put up by treatments, and, like "satellite navigation", find alternative routes by which to carry on their "journey" to cancer progression.
Collapse
Affiliation(s)
| | - Athina-Myrto Chioni
- School of Life Sciences Pharmacy and Chemistry, Biomolecular Sciences Department, Kingston University London, Kingston-upon-Thames KT1 2EE, UK;
| |
Collapse
|
12
|
Babar Q, Saeed A, Tabish TA, Sarwar M, Thorat ND. Targeting the tumor microenvironment: Potential strategy for cancer therapeutics. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166746. [PMID: 37160171 DOI: 10.1016/j.bbadis.2023.166746] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/30/2023] [Accepted: 05/02/2023] [Indexed: 05/11/2023]
Abstract
Cellular and stromal components including tumor cells, immune cells, mesenchymal cells, cancer-linked fibroblasts, and extracellular matrix, constituent tumor microenvironment (TME). TME plays a crucial role in reprogramming tumor initiation, uncontrolled proliferation, invasion and metastasis as well as response to therapeutic modalities. In recent years targeting the TME has developed as a potential strategy for treatment of cancer because of its life-threatening functions in restricting tumor development and modulating responses to standard-of-care medicines. Cold atmospheric plasma, oncolytic viral therapy, bacterial therapy, nano-vaccine, and repurposed pharmaceuticals with combination therapy, antiangiogenic drugs, and immunotherapies are among the most effective therapies directed by TME that have either been clinically authorized or are currently being studied. This article discusses above-mentioned therapies in light of targeting TME. We also cover problems related to the TME-targeted therapies, as well as future insights and practical uses in this rapidly growing field.
Collapse
Affiliation(s)
- Quratulain Babar
- Department of Biochemistry Government College University, Faisalabad, Pakistan
| | - Ayesha Saeed
- Department of Biochemistry Government College University, Faisalabad, Pakistan
| | - Tanveer A Tabish
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Mohsin Sarwar
- Department of Biochemistry University of Management and Technology, Lahore, Pakistan
| | - Nanasaheb D Thorat
- Department of Physics, Bernal Institute, Castletroy, Limerick V94T9PX, Ireland; Nuffield Department of Women's and Reproductive Health, John Radcliffe Hospital, Medical Sciences Division, University of Oxford, Oxford OX3 9DU, United Kingdom; Limerick Digital Cancer Research Centre (LDCRC) University of Limerick, Castletroy, Limerick V94T9PX, Ireland.
| |
Collapse
|
13
|
Hu H, Li B, Wang J, Tan Y, Xu M, Xu W, Lu H. New advances into cisplatin resistance in head and neck squamous carcinoma: Mechanisms and therapeutic aspects. Biomed Pharmacother 2023; 163:114778. [PMID: 37137185 DOI: 10.1016/j.biopha.2023.114778] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/20/2023] [Accepted: 04/23/2023] [Indexed: 05/05/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) arises from the interplay of multiple factors, such as smoking, alcohol consumption, and viral infections. Cisplatin-based concurrent radiotherapy regimens represent the first-line treatment for advanced HNSCC cases. However, cisplatin resistance significantly contributes to poor prognoses in HNSCC patients, making it crucial to unravel the underlying mechanisms to overcome this resistance. The complexity of cisplatin resistance in HNSCC involves cancer stem cells, autophagy, epithelial-mesenchymal transition, drug efflux, and metabolic reprogramming. Recent advances in nanodrug delivery systems, combined with existing small-molecule inhibitors and innovative genetic technologies, have opened new therapeutic avenues for addressing cisplatin resistance in HNSCC. This review systematically summarizes research progress from the past five years on cisplatin resistance in HNSCC, with a particular focus on the roles of cancer stem cells and autophagy. Additionally, potential future treatment strategies to overcome cisplatin resistance are discussed, including the targeting of cancer stem cells or autophagy through nanoparticle-based drug delivery systems. Furthermore, the review highlights the prospects and challenges associated with nanodelivery platforms in addressing cisplatin resistance in HNSCC.
Collapse
Affiliation(s)
- Hanlin Hu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Bo Li
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Junke Wang
- Department of Cardiology, Qingdao Hiser Hospital Affiliated to Qingdao University, Qingdao, China.
| | - Ye Tan
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Mingjin Xu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Wenhua Xu
- Institute of Regenerative Medicine and Laboratory Technology Innovation, Qingdao University, Qingdao, China.
| | - Haijun Lu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
14
|
Audero MM, Carvalho TMA, Ruffinatti FA, Loeck T, Yassine M, Chinigò G, Folcher A, Farfariello V, Amadori S, Vaghi C, Schwab A, Reshkin SJ, Cardone RA, Prevarskaya N, Fiorio Pla A. Acidic Growth Conditions Promote Epithelial-to-Mesenchymal Transition to Select More Aggressive PDAC Cell Phenotypes In Vitro. Cancers (Basel) 2023; 15:cancers15092572. [PMID: 37174038 PMCID: PMC10177299 DOI: 10.3390/cancers15092572] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/28/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is characterized by an acidic microenvironment, which contributes to therapeutic failure. So far there is a lack of knowledge with respect to the role of the acidic microenvironment in the invasive process. This work aimed to study the phenotypic and genetic response of PDAC cells to acidic stress along the different stages of selection. To this end, we subjected the cells to short- and long-term acidic pressure and recovery to pHe 7.4. This treatment aimed at mimicking PDAC edges and consequent cancer cell escape from the tumor. The impact of acidosis was assessed for cell morphology, proliferation, adhesion, migration, invasion, and epithelial-mesenchymal transition (EMT) via functional in vitro assays and RNA sequencing. Our results indicate that short acidic treatment limits growth, adhesion, invasion, and viability of PDAC cells. As the acid treatment progresses, it selects cancer cells with enhanced migration and invasion abilities induced by EMT, potentiating their metastatic potential when re-exposed to pHe 7.4. The RNA-seq analysis of PANC-1 cells exposed to short-term acidosis and pHe-selected recovered to pHe 7.4 revealed distinct transcriptome rewiring. We describe an enrichment of genes relevant to proliferation, migration, EMT, and invasion in acid-selected cells. Our work clearly demonstrates that upon acidosis stress, PDAC cells acquire more invasive cell phenotypes by promoting EMT and thus paving the way for more aggressive cell phenotypes.
Collapse
Affiliation(s)
- Madelaine Magalì Audero
- U1003-PHYCELL-Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d'Ascq, 59000 Lille, France
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| | | | - Federico Alessandro Ruffinatti
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| | - Thorsten Loeck
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Maya Yassine
- U1003-PHYCELL-Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d'Ascq, 59000 Lille, France
| | - Giorgia Chinigò
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| | - Antoine Folcher
- U1003-PHYCELL-Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d'Ascq, 59000 Lille, France
| | - Valerio Farfariello
- U1003-PHYCELL-Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d'Ascq, 59000 Lille, France
| | - Samuele Amadori
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| | - Chiara Vaghi
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| | - Albrecht Schwab
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Stephan J Reshkin
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70126 Bari, Italy
| | - Rosa Angela Cardone
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70126 Bari, Italy
| | - Natalia Prevarskaya
- U1003-PHYCELL-Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d'Ascq, 59000 Lille, France
| | - Alessandra Fiorio Pla
- U1003-PHYCELL-Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d'Ascq, 59000 Lille, France
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| |
Collapse
|
15
|
Huni KC, Cheung J, Sullivan M, Robison WT, Howard KM, Kingsley K. Chemotherapeutic Drug Resistance Associated with Differential miRNA Expression of miR-375 and miR-27 among Oral Cancer Cell Lines. Int J Mol Sci 2023; 24:ijms24021244. [PMID: 36674758 PMCID: PMC9865318 DOI: 10.3390/ijms24021244] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/01/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Recent advances have suggested that non-coding miRNAs (such as miR-21, miR-27, miR-145, miR-155, miR-365, miR-375 and miR-494) may be involved in multiple aspects of oral cancer chemotherapeutic responsiveness. This study evaluated whether these specific miRNAs are correlated with oral cancer responsiveness to chemotherapies, including Paclitaxel, Cisplatin and Fluorouracil (5FU). Commercially available and well-characterized oral squamous cell carcinoma cell lines (SCC4, SCC9, SCC15, SCC25 and CAL27) revealed differing resistance and chemosensitivity to these agents-with SCC9 and SCC25 demonstrating the most resistance to all chemotherapeutic agents. SCC9 and SCC25 were also the only cell lines that expressed miR-375, and were the only cell lines that did not express miR-27. In addition, the expression of miR-375 was associated with the upregulation of Rearranged L-myc fusion (RLF) and the downregulation of Centriolar protein B (POC1), whereas lack of miR-27 expression was associated with Nucleophosmin 1 (NPM1) expression. These data have revealed important regulatory pathways and mechanisms associated with oral cancer proliferation and resistance that must be explored in future studies of potential therapeutic interventions.
Collapse
Affiliation(s)
- Kieran Caberto Huni
- Department of Advanced Education in Orthodontic Dentistry, School of Dental Medicine, University of Nevada-Las Vegas, 1700 W. Charleston Boulevard, Las Vegas, NV 89106, USA
| | - Jacky Cheung
- Department of Clinical Sciences, School of Dental Medicine, University of Nevada-Las Vegas, 1700 W. Charleston Boulevard, Las Vegas, NV 89106, USA
| | - Madeline Sullivan
- Department of Clinical Sciences, School of Dental Medicine, University of Nevada-Las Vegas, 1700 W. Charleston Boulevard, Las Vegas, NV 89106, USA
| | - William Taylor Robison
- Department of Clinical Sciences, School of Dental Medicine, University of Nevada-Las Vegas, 1700 W. Charleston Boulevard, Las Vegas, NV 89106, USA
| | - Katherine M. Howard
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada-Las Vegas, 1001 Shadow Lane, Las Vegas, NV 89106, USA
| | - Karl Kingsley
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada-Las Vegas, 1001 Shadow Lane, Las Vegas, NV 89106, USA
- Correspondence: ; Tel.: +1-702-774-2623
| |
Collapse
|
16
|
Peppicelli S, Ruzzolini J, Lulli M, Biagioni A, Bianchini F, Caldarella A, Nediani C, Andreucci E, Calorini L. Extracellular Acidosis Differentially Regulates Estrogen Receptor β-Dependent EMT Reprogramming in Female and Male Melanoma Cells. Int J Mol Sci 2022; 23:15374. [PMID: 36499700 PMCID: PMC9736857 DOI: 10.3390/ijms232315374] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/16/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Clinical outcomes of melanoma patients pointed out a gender disparity that supports a correlation between sex hormone activity on estrogen receptors (ER) and melanoma development and progression. Here, we found that the epithelial-to-mesenchymal transition (EMT) of melanoma cells induced by extracellular acidosis, which is a crucial hallmark of solid cancers, correlates with the expression of ERβ, the most representative ER on melanoma cells. Extracellular acidosis induces an enhanced expression of ERβ in female cells and EMT markers remain unchanged, while extracellular acidosis did not induce the expression of ERβ in male cells and EMT was strongly promoted. An inverse relationship between ERβ expression and EMT markers in melanoma cells of different sex exposed to extracellular acidosis was revealed by two different technical approaches: florescence-activated cell sorting of high ERβ expressing cell subpopulations and ERβ receptor silencing. Finally, we found that ERβ regulates EMT through NF-κB activation. These results demonstrate that extracellular acidosis drives a differential ERβ regulation in male and female melanoma cells and that this gender disparity might open new perspectives for personalized therapeutic approaches.
Collapse
Affiliation(s)
- Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Jessica Ruzzolini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Adele Caldarella
- Tuscany Cancer Registry, Clinical Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO)-Florence, 50139 Florence, Italy
| | - Chiara Nediani
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Elena Andreucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Lido Calorini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
- Center of Excellence for Research, Transfer and High Education DenoTHE, University of Florence, 50134 Florence, Italy
| |
Collapse
|
17
|
Acid Adaptation Promotes TRPC1 Plasma Membrane Localization Leading to Pancreatic Ductal Adenocarcinoma Cell Proliferation and Migration through Ca 2+ Entry and Interaction with PI3K/CaM. Cancers (Basel) 2022; 14:cancers14194946. [PMID: 36230869 PMCID: PMC9563726 DOI: 10.3390/cancers14194946] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/03/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers globally, with a 5-year overall survival of less than 10%. The development and progression of PDAC are linked to its fluctuating acidic tumor microenvironment. Ion channels act as important sensors of this acidic tumor microenvironment. They transduce extracellular signals and regulate signaling pathways involved in all hallmarks of cancer. In this study, we evaluated the interplay between a pH-sensitive ion channel, the calcium (Ca2+) channel transient receptor potential C1 (TRPC1), and three different stages of the tumor microenvironment, normal pH, acid adaptation, and acid recovery, and its impact on PDAC cell migration, proliferation, and cell cycle progression. In acid adaptation and recovery conditions, TRPC1 localizes to the plasma membrane, where it interacts with PI3K and calmodulin, and permits Ca2+ entry, which results in downstream signaling, leading to proliferation and migration. Thus, TRPC1 exerts a more aggressive role after adaptation to the acidic tumor microenvironment. Abstract Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal malignancies, with a low overall survival rate of less than 10% and limited therapeutic options. Fluctuations in tumor microenvironment pH are a hallmark of PDAC development and progression. Many ion channels are bona fide cellular sensors of changes in pH. Yet, the interplay between the acidic tumor microenvironment and ion channel regulation in PDAC is poorly understood. In this study, we show that acid adaption increases PANC-1 cell migration but attenuates proliferation and spheroid growth, which are restored upon recovery. Moreover, acid adaptation and recovery conditions favor the plasma membrane localization of the pH-sensitive calcium (Ca2+) channel transient receptor potential C1 (TRPC1), TRPC1-mediated Ca2+ influx, channel interaction with the PI3K p85α subunit and calmodulin (CaM), and AKT and ERK1/2 activation. Knockdown (KD) of TRPC1 suppresses cell migration, proliferation, and spheroid growth, notably in acid-recovered cells. KD of TRPC1 causes the accumulation of cells in G0/G1 and G2/M phases, along with reduced expression of CDK6, −2, and −1, and cyclin A, and increased expression of p21CIP1. TRPC1 silencing decreases the basal Ca2+ influx in acid-adapted and -recovered cells, but not in normal pH conditions, and Ca2+ chelation reduces cell migration and proliferation solely in acid adaptation and recovery conditions. In conclusion, acid adaptation and recovery reinforce the involvement of TRPC1 in migration, proliferation, and cell cycle progression by permitting Ca2+ entry and forming a complex with the PI3K p85α subunit and CaM.
Collapse
|