1
|
Han L, Sun Y, Yang K, Long C. Inhibition of JAK2/STAT3 by pacritinib synergizes with chemotherapy in esophageal carcinoma. Toxicol In Vitro 2025; 106:106056. [PMID: 40112935 DOI: 10.1016/j.tiv.2025.106056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
The poor outcomes associated with esophageal carcinoma, particularly in advanced stages, necessitate the development of new treatment strategies. This study examines the efficacy of pacritinib, a multi-kinase inhibitor, both alone and in combination with carboplatin, in preclinical esophageal carcinoma models. Six esophageal carcinoma cell lines (KYSE-70, OE33, FLO-1, KYAE-1, ESO 26, and HCE-6) were treated with pacritinib, resulting in a dose-dependent reduction in cell viability. Combination index (CI) analysis demonstrated strong synergy between pacritinib and carboplatin across this panel of cell lines. In in vivo esophageal carcinoma xenograft model, pacritinib alone significantly reduced tumor growth and improved survival rates compared to control. Notably, the combination of pacritinib and carboplatin further reduced tumor growth and improved survival rates compared to either treatment alone. Toxicity assessment showed that neither single-agent nor combination treatment resulted in significantly altered levels of body weight and serum markers, supporting the safety profile of pacritinib in combination with carboplatin. Mechanistic studies revealed that while pacritinib inhibited the phosphorylation of JAK, STAT3, and IRAK1 in esophageal carcinoma cells, it is the suppression of the JAK/STAT3 pathway, rather than IRAK1, that is responsible for the synergistic effect with carboplatin. Our findings indicate that pacritinib possesses potent anti-tumor activity in esophageal carcinoma and enhances the efficacy of carboplatin through the suppression of JAK/STAT3 signaling, warranting further clinical investigation.
Collapse
Affiliation(s)
- Lan Han
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Yinyin Sun
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Kai Yang
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China.
| | - Cheng Long
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China.
| |
Collapse
|
2
|
Xiao ZW, Zeng YC, Ji LT, Yuan JT, Li L. Nitric oxide synthase 1 inhibits the progression of esophageal cancer through interacting with nitric oxide synthase 1 adaptor protein. World J Gastrointest Oncol 2025; 17:103843. [PMID: 40235872 PMCID: PMC11995332 DOI: 10.4251/wjgo.v17.i4.103843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/22/2025] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Esophageal cancer (ESCA) is among the most prevalent and lethal tumors globally. While nitric oxide synthase 1 (NOS1) is recognized for its important involvement in various cancers, its specific function in ESCA remains unclear. AIM To explore the potential role and underlying mechanisms of NOS1 in ESCA. METHODS Survival rates were analyzed using GeneCards and Gene Expression Profiling Interactive Analysis. The effects and mechanisms of NOS1 on ESCA cells were evaluated via the Cell Counting Kit-8 assay, scratch assay, Transwell assay, flow cytometry, quantitative polymerase chain reaction, western blotting, and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick end labeling staining. The protein interaction network was used to screen the interacting proteins of NOS1 and validate these interactions through co-immunoprecipitation and dual luciferase assays. Additionally, a nude mouse xenograft model was established to evaluate the effect of NOS1 in vivo. RESULTS The survival rate of patients with ESCA with high NOS1 expression was higher than that of patients with low NOS1 expression. NOS1 expression in ESCA cell lines was lower than that in normal esophageal epithelial cells. Overexpression of NOS1 (oe-NOS1) inhibited proliferation, invasion, and migration abilities in ESCA cell lines, resulting in decreased autophagy levels and increased apoptosis, pyroptosis, and ferroptosis. Protein interaction studies confirmed the interaction between NOS1 and NOS1 adaptor protein (NOS1AP). Following oe-NOS1 and the silencing of NOS1AP, levels of P62 and microtubule-associated protein 1 light chain 3 beta increased both in vitro and in vivo. Furthermore, the expression levels of E-cadherin, along with the activation of phosphatidylinositol 3-kinase (PI3K) and protein kinase B (AKT), were inhibited in ESCA cell lines. CONCLUSION NOS1 and NOS1 proteins interact to suppress autophagy, activate the PI3K/AKT pathway, and exert anti-cancer effects in ESCA.
Collapse
Affiliation(s)
- Zi-Wei Xiao
- College of Medical, Hunan Normal University, Changsha 410081, Hunan Province, China
| | - Ying-Chao Zeng
- College of Medical, Hunan Normal University, Changsha 410081, Hunan Province, China
| | - Lin-Tao Ji
- College of Medical, Hunan Normal University, Changsha 410081, Hunan Province, China
| | - Jia-Tao Yuan
- College of Medical, Hunan Normal University, Changsha 410081, Hunan Province, China
| | - Lin Li
- College of Medical, Hunan Normal University, Changsha 410081, Hunan Province, China
| |
Collapse
|
3
|
Dasharathy S, Pranay, Devadas SK, Tripathi E, Karyala P. Emerging role of deubiquitinases in modulating cancer chemoresistance. Drug Discov Today 2025; 30:104339. [PMID: 40118446 DOI: 10.1016/j.drudis.2025.104339] [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: 01/11/2025] [Revised: 03/08/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
Chemotherapy remains a gold standard in cancer treatment by targeting the rapidly dividing cancer cells. However, chemoresistance is a major obstacle to successful cancer treatment, often leading to recurrence, metastasis, and high mortality. Deubiquitinases (DUBs), enzymes that remove ubiquitin and stabilize proteins, have been implicated in chemoresistance and can either promote therapeutic resistance or enhance sensitivity depending on their targets. In this review, we highlight the chemoresistance mechanisms of DUBs in various cancers, including breast, lung, liver, gastrointestinal, colorectal, ovarian, prostate, and blood cancers. Given these mechanisms, the development of DUB inhibitors has gained considerable attention in cancer therapeutics and combination therapies involving these inhibitors show potential to overcome drug resistance and improving treatment outcomes.
Collapse
Affiliation(s)
- Sukeerthi Dasharathy
- Department of Biotechnology, Faculty of Life and Allied Health Sciences, M.S. Ramaiah University of Applied Sciences, Bangalore 560054, India
| | - Pranay
- Department of Biotechnology, Faculty of Life and Allied Health Sciences, M.S. Ramaiah University of Applied Sciences, Bangalore 560054, India
| | - Santhosh K Devadas
- Department of Medical Oncology, Ramaiah Medical College and Hospital, M.S. Ramaiah University of Applied Sciences, Bangalore 560054, India
| | - Ekta Tripathi
- Department of Biotechnology, Faculty of Life and Allied Health Sciences, M.S. Ramaiah University of Applied Sciences, Bangalore 560054, India.
| | - Prashanthi Karyala
- Department of Biotechnology, Faculty of Life and Allied Health Sciences, M.S. Ramaiah University of Applied Sciences, Bangalore 560054, India.
| |
Collapse
|
4
|
Li Y, Guo Q, Wang H, Wang R, Kang W, Zhang CL, Zhang D, Xiao K, Sun Z. Associations between vascular endothelial growth factor polymorphisms and response to 5-FU-based pharmaceutical therapy in esophageal squamous cell carcinoma: A meta-analysis. TUMORI JOURNAL 2025; 111:112-120. [PMID: 40012106 DOI: 10.1177/03008916251322057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
BACKGROUND AND AIMS Vascular endothelial growth factor (VEGF) gene polymorphisms are associated with the response to pharmaceutical therapy in many cancers. This study aimed to investigate the effects of VEGF gene polymorphisms in esophageal squamous cell carcinoma patients receiving pharmaceutical therapy. METHODS This literature-based meta-analysis was performed with keywords related to VEGF gene polymorphisms and clinical response in esophageal squamous carcinoma patients receiving pharmaceutical therapy (including 5-FU, cisplatin, oxaliplatin, and calcium folinate). After a series of bias grading analyses and DerSimonian-Laird method analysis, odds ratios and 95% confidence intervals were calculated to examine the potential relationships. Sensitivity and subgroup analyses were subsequently performed to determine the major causes of heterogeneity. RESULTS Heterogeneity was dramatically reduced after the removal of one study from the analysis (I2 = 37%, P = 0.19). The remaining studies involved 5-FU-based treatment. The presence of VEGF G-1154A and VEGF-634C/G was found to be correlated with patient response to 5-FU/CDDP-based treatment, whereas VEGF-2549I/D was correlated with response to 5-FU/oxaliplatin-based treatment, and VEGF-936C/T was associated with both 5-FU/CDDP- and 5-FU/oxaliplatin-based treatment response. CONCLUSION VEGF gene polymorphisms affect the response of esophageal squamous carcinoma patients receiving pharmaceutical therapy, especially 5-FU-based treatments.
Collapse
Affiliation(s)
- Yonghui Li
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, P. R. China
| | - Qiang Guo
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, P. R. China
| | - Haibo Wang
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, P. R. China
| | - Ruiyao Wang
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, P. R. China
| | - WenLi Kang
- Department of Obstetrical, Affiliated Hospital of Hebei University, Baoding, Hebei, P. R. China
| | - Cheng Long Zhang
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, P. R. China
| | - Duo Zhang
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, P. R. China
| | - Kuo Xiao
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, P. R. China
| | - ZhenQing Sun
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei, P. R. China
| |
Collapse
|
5
|
Lahouty M, Soleymanzadeh A, Kazemi S, Saadati-Maleki H, Masoudi S, Ghasemi A, Kazemi T, Mehranfar S, Fadaee M. Cell-based immunotherapy in oesophageal cancer. J Drug Target 2025:1-11. [PMID: 40063049 DOI: 10.1080/1061186x.2025.2477077] [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: 01/21/2025] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 03/18/2025]
Abstract
Oesophageal cancer (EC) is among the most common illnesses globally, and its prognosis is unfavourable. Surgery, radiotherapy and chemotherapy are the primary therapy options for EC. Despite the occasional efficacy of these traditional treatment modalities, individuals with EC remain at a significant risk for local recurrence and metastasis. Consequently, innovative and efficacious medicines are required. In recent decades, clinical practice has effectively implemented cell therapy, which includes both stem cell and non-stem cell-based approaches, as an innovative tumour treatment, offering renewed hope to patients with oesophageal squamous cell carcinoma (ESCC). This paper examines the theoretical framework and contemporary advancements in cell treatment for individuals with EC. We further described current clinical studies and summarised essential data related to survival and safety assessments.
Collapse
Affiliation(s)
- Masoud Lahouty
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Sama Kazemi
- Faculty of Medicine, Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Haniyeh Saadati-Maleki
- Faculty of Medicine, Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Sanaz Masoudi
- Faculty of Medicine, Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Arash Ghasemi
- Faculty of Medicine, Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Mehranfar
- Department of Genetics and Immunology, Urmia University of Medical Sciences, Urmia, Iran
| | - Manouchehr Fadaee
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Liu Z, Liu J, Wu Y, Zhou Z, Ousmane D, Zeinalzadeh Z, Wang J. Shared chemoresistance genes in ESCC and cervical Cancer: Insights from pharmacogenomics and Mendelian randomization. Int Immunopharmacol 2025; 147:113933. [PMID: 39755112 DOI: 10.1016/j.intimp.2024.113933] [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: 03/15/2024] [Revised: 10/29/2024] [Accepted: 12/21/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND Neoadjuvant chemotherapy, particularly the use of platinum-based compounds and taxanes, is pivotal in the treatment of epithelial-derived tumors, such as cervical cancer and esophageal squamous cell carcinoma (ESCC); however, resistance remains a significant challenge. Utilizing Mendelian randomization (MR) with pharmacogenomics offers a novel approach to understanding the genetic underpinnings of drug responses, thereby aiding in personalized treatment. METHODS Single-cell RNA sequencing (scRNA-seq) analysis revealed a shared cellular subpopulation of CD8 + T effector memory (CD8 + TEM) cells that are pivotal in mediating chemotherapy resistance in ESCC and cervical cancer. A two-sample approach was employed for MR using data from genome-wide association studies, focusing on single nucleotide polymorphisms (SNPs) linked to CD8 + TEM cell expression. The SNPs were carefully selected, and statistical models, including the Wald ratio and inverse variance weighted methods, were used for robust causal effect estimation. These were supplemented by MR-Egger and weighted median analyses to address pleiotropy and variant heterogeneity. 3-(4,5-Dimethylthiazol-2-yl)-2,5 diphenyl tetrazolium bromide (MTT) and immunohistochemistry assays were used to verify the relationship between the gene and drug sensitivity. RESULTS Increased proportion of CD8 + TEM cells were observed in resistant samples. MR identified IL32, SPOCK1, and TRBC2 as key genes associated with resistance to cisplatin, carboplatin, and paclitaxel, respectively. These findings were validated across various cohorts and underscored the role of CD8 + TEM cells in drug responsiveness. The results of the MTT and immunohistochemistry assays confirmed the MR findings. CONCLUSIONS Our study highlights the significant role of CD8 + TEM cells in the chemoresistance of ESCC and cervical cancer and identified three genetic markers crucial for resistance to common chemotherapeutic agents. These findings suggest potential pathways for developing personalized treatment strategies, offering clinically relevant insights that could enhance therapeutic efficacy and help overcome drug resistance in patients with ESCC or cervical cancer.
Collapse
Affiliation(s)
- Ziyu Liu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathology, School of Basic Medicine, Central South University, Changsha City, Hunan Province, China; FuRong Laboratory, Changsha City, Hunan Province, China
| | - Jie Liu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathology, School of Basic Medicine, Central South University, Changsha City, Hunan Province, China
| | - Yanhao Wu
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Zongjiang Zhou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathology, School of Basic Medicine, Central South University, Changsha City, Hunan Province, China
| | - Diabate Ousmane
- Department of Pathology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathology, School of Basic Medicine, Central South University, Changsha City, Hunan Province, China
| | - Zahra Zeinalzadeh
- Department of Pathology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathology, School of Basic Medicine, Central South University, Changsha City, Hunan Province, China
| | - Junpu Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathology, School of Basic Medicine, Central South University, Changsha City, Hunan Province, China; Ultrapathology (Biomedical Electron Microscopy) Center, Department of Pathology, Xiang-ya Hospital, Central South University, Changsha City, Hunan Province, China; Key Laboratory of Hunan Province in Neurodegenerative Disorders, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; FuRong Laboratory, Changsha City, Hunan Province, China.
| |
Collapse
|
7
|
Dong XM, Chen L, Xu YX, Wu P, Xie T, Liu ZQ. Exploring metabolic reprogramming in esophageal cancer: the role of key enzymes in glucose, amino acid, and nucleotide pathways and targeted therapies. Cancer Gene Ther 2025; 32:165-183. [PMID: 39794467 DOI: 10.1038/s41417-024-00858-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 01/13/2025]
Abstract
Esophageal cancer (EC) is one of the most common malignancies worldwide with the character of poor prognosis and high mortality. Despite significant advancements have been achieved in elucidating the molecular mechanisms of EC, for example, in the discovery of new biomarkers and metabolic pathways, effective treatment options for patients with advanced EC are still limited. Metabolic heterogeneity in EC is a critical factor contributing to poor clinical outcomes. This heterogeneity arises from the complex interplay between the tumor microenvironment and genetic factors of tumor cells, which drives significant metabolic alterations in EC, a process known as metabolic reprogramming. Understanding the mechanisms of metabolic reprogramming is essential for developing new antitumor therapies and improving treatment outcomes. Targeting the distinct metabolic alterations in EC could enable more precise and effective therapies. In this review, we explore the complex metabolic changes in glucose, amino acid, and nucleotide metabolism during the progression of EC, and how these changes drive unique nutritional demands in cancer cells. We also evaluate potential therapies targeting key metabolic enzymes and their clinical applicability. Our work will contribute to enhancing knowledge of metabolic reprogramming in EC and provide new insights and approaches for the clinical treatment of EC.
Collapse
Affiliation(s)
- Xue-Man Dong
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Lin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Yu-Xin Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Pu Wu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.
| | - Zhao-Qian Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.
| |
Collapse
|
8
|
Han H, Li Y, Lin Z, Ma X, Huang W, Lu C, Ma R, Han R. Exosomal miR-130a-3p confers cisplatin resistance in esophageal cancer by regulating ferroptosis via the suppression of METTL14-mediated m6A RNA methylation of FSP1. Int Immunopharmacol 2025; 146:113804. [PMID: 39689599 DOI: 10.1016/j.intimp.2024.113804] [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: 10/13/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024]
Abstract
Exosomal microRNA (miRNA)s have been proven to affect recipient cell chemoresistance in several cancers. This research attempted to uncover the role of exosomal miRNA and the regulatory mechanism in cisplatin resistance of esophageal cancer (EC). Cisplatin-resistant EC cells (KYSE-150-CisR and TE-1-CisR) were established by the parental cells (KYSE-150 and TE-1) treated with a gradual increase of cisplatin concentration. Exosomes from both cisplatin-resistant EC cells and the parental one were obtained with ultracentrifugation (CisR-exo and CisS-exo), and identified by transmission electron microscopy and nanoparticle tracking analysis. The effect of CisR-exo on the cisplatin resistance of EC was assessed by in vitro (and in vivo functional experiments.Intracellular reactive oxygen species and iron were determined by the corresponding kits. The m6A dot blot assay and methylated RIP-qPCR was conducted to analyze the m6A modification level. Dual-luciferase reporter assay was performed to confirm the intermolecular interaction. Increased levels of miR-130a-3p were observed in both KYSE-150CisR and TE-1CisR cells, as well as their derived CisR-exos when compared with the parental cells and CisS-exos. Exosomal miR-130a-3p from cisplatin-resistant EC cells conferred cisplatin resistance to EC in vitro and in vivo, which might be mediated by the suppression of ferroptosis. Mechanically, KYSE-150CisR derived exosomal miR-130a-3p interacted with METTL14 to inhibit FSP1 (a ferroptosis suppressor) m6A modification of recipient cells. Overexpressing METTL14 restrained the cisplatin resistance disseminated by CisR-exos in KYSE-150 cells. Cisplatin-resistant EC cell-isolated exosomal miR-130a-3p suppressed the m6A modification of FSP1 to modulate ferroptosis, enhancing cisplatin resistance.
Collapse
Affiliation(s)
- Hu Han
- Department of Oncology, The First Affiliated Hospital of Shihezi University, Shihezi 832008, China.
| | - Yan Li
- Medical Department, The First Affiliated Hospital of Shihezi University, Shihezi 832008, China.
| | - Zhiyi Lin
- Department of Oncology, The First Affiliated Hospital of Shihezi University, Shihezi 832008, China.
| | - Xiaoping Ma
- Department of Oncology, The First Affiliated Hospital of Shihezi University, Shihezi 832008, China.
| | - Wukui Huang
- Department of Interventional Diagnosis and Treatment, Affiliated Cancer Hospital, Xinjiang Medical University, Urumqi 830054, China.
| | - Cengceng Lu
- Department of Oncology, The First Affiliated Hospital of Shihezi University, Shihezi 832008, China.
| | - Rongyan Ma
- Department of Oncology, The First Affiliated Hospital of Shihezi University, Shihezi 832008, China.
| | - Rui Han
- Medical Department, The First Affiliated Hospital of Shihezi University, Shihezi 832008, China.
| |
Collapse
|
9
|
Li DD, Jin JC, Liu XW, Liu SY, Ji FJ, Liu T. Nanoparticle delivery of si-Notch1 modulates metabolic reprogramming to affect 5-FU resistance and cell pyroptosis in colorectal cancer. Cancer Nanotechnol 2024; 15:23. [DOI: 10.1186/s12645-024-00259-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 03/29/2024] [Indexed: 01/05/2025] Open
Abstract
Abstract
Background
Nanocarrier delivery of small interfering RNAs (siRNAs) to silence cancer-associated genes is a promising method for cancer treatment. Here, we explored the role and mechanisms of PLAG NPs-delivered si-Notch1 in colorectal cancer (CRC).
Results
High Notch1 expression was observed in both sensitive and resistant CRC tissues and cells. Notch1 silencing repressed proliferation and facilitates apoptosis of resistant CRC cells, and suppressed glycolysis and promoted pyroptosis in resistant CRC cells. Notch1 directly interacts with PCAF. Notch1 knockdown’s suppressive effect on glycolysis was reversed by overexpression of PCAF. Moreover, a nanocarrier called PLAG NPs was built with a higher delivery efficiency compared with lipo2000. Si-Notch1 delivered by PLAG NPs efficiently overcame the CRC cells’ 5-FU resistance and facilitated pyroptosis in a CRC mouse model.
Conclusions
PLAG NPs carrying si-Notch1 had a great advantage in the extension of half-life circulation and targeting ability, providing a theoretical foundation for precise clinical treatment of CRC.
Collapse
|
10
|
Herms A, Fernandez-Antoran D, Alcolea MP, Kalogeropoulou A, Banerjee U, Piedrafita G, Abby E, Valverde-Lopez JA, Ferreira IS, Caseda I, Bejar MT, Dentro SC, Vidal-Notari S, Ong SH, Colom B, Murai K, King C, Mahbubani K, Saeb-Parsy K, Lowe AR, Gerstung M, Jones PH. Self-sustaining long-term 3D epithelioid cultures reveal drivers of clonal expansion in esophageal epithelium. Nat Genet 2024; 56:2158-2173. [PMID: 39313617 PMCID: PMC11525200 DOI: 10.1038/s41588-024-01875-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 07/18/2024] [Indexed: 09/25/2024]
Abstract
Aging epithelia are colonized by somatic mutations, which are subjected to selection influenced by intrinsic and extrinsic factors. The lack of suitable culture systems has slowed the study of this and other long-term biological processes. Here, we describe epithelioids, a facile, cost-effective method of culturing multiple mouse and human epithelia. Esophageal epithelioids self-maintain without passaging for at least 1 year, maintaining a three-dimensional structure with proliferative basal cells that differentiate into suprabasal cells, which eventually shed and retain genomic stability. Live imaging over 5 months showed that epithelioids replicate in vivo cell dynamics. Epithelioids support genetic manipulation and enable the study of mutant cell competition and selection in three-dimensional epithelia, and show how anti-cancer treatments modulate competition between transformed and wild-type cells. Finally, a targeted CRISPR-Cas9 screen shows that epithelioids recapitulate mutant gene selection in aging human esophagus and identifies additional drivers of clonal expansion, resolving the genetic networks underpinning competitive fitness.
Collapse
Affiliation(s)
- Albert Herms
- Wellcome Sanger Institute, Hinxton, UK
- Department of Biomedical Sciences, Universitat de Barcelona, Barcelona, Spain
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - David Fernandez-Antoran
- Wellcome Sanger Institute, Hinxton, UK
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
- ARAID Foundation, Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain
| | - Maria P Alcolea
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | | | | | - Gabriel Piedrafita
- Wellcome Sanger Institute, Hinxton, UK
- Department of Biochemistry and Molecular Biology, Complutense University of Madrid, Madrid, Spain
- Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | | | - Inês S Ferreira
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
| | - Irene Caseda
- Department of Biomedical Sciences, Universitat de Barcelona, Barcelona, Spain
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Maria T Bejar
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Stefan C Dentro
- Wellcome Sanger Institute, Hinxton, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK
- Artificial Intelligence in Oncology (B450), Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Sara Vidal-Notari
- Department of Biochemistry and Molecular Biology, Complutense University of Madrid, Madrid, Spain
- Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Bartomeu Colom
- Wellcome Sanger Institute, Hinxton, UK
- Cambridge Institute of Science, Altos Labs, Cambridge, UK
| | | | | | - Krishnaa Mahbubani
- Department of Surgery, University of Cambridge, Cambridge, UK
- Collaborative Biorepository for Translational Medicine (CBTM), Cambridge NIHR Biomedical Research Centre, Cambridge, UK
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, Cambridge, UK
- Collaborative Biorepository for Translational Medicine (CBTM), Cambridge NIHR Biomedical Research Centre, Cambridge, UK
| | - Alan R Lowe
- Institute for Structural and Molecular Biology, University College London, London, UK
- Institute for the Physics of Living Systems, University College London, London, UK
- Department of Physics and Astronomy, University College London, London, UK
| | - Moritz Gerstung
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK
- Artificial Intelligence in Oncology (B450), Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | - Philip H Jones
- Wellcome Sanger Institute, Hinxton, UK.
- Department of Oncology, Hutchison Research Centre, University of Cambridge, Cambridge, UK.
| |
Collapse
|
11
|
Kai J, Kang K, Jiang Z, Xiong F, Wang S. Capivasertib reverses chemotherapy-induced esophageal cancer resistance via inhibiting Akt-associated Mcl-1 upregulation. Heliyon 2024; 10:e33567. [PMID: 39050467 PMCID: PMC11266993 DOI: 10.1016/j.heliyon.2024.e33567] [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: 02/04/2024] [Revised: 06/22/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
The development of resistance to chemotherapy in esophageal cancer represents a significant challenge in cancer treatment. Therefore, our study aimed to identify effective therapeutic strategies by examining the molecules involved in this chemoresistance. We consistently observed an increase in the expression of Mcl-1 in cells exposed to both short and long-term treatment with cisplatin, a drug commonly used in esophageal cancer therapy. Functional analysis showed that Mcl-1 regulates esophageal cancer cell response to cisplatin treatment. Notably, this upregulation of Mcl-1 was not dependent on eukaryotic initiation factor 4E (eIF4E). Instead, it was associated with increased stability due to the activation of Akt. Capivasertib, a potent pan-Akt kinase drug, significantly decreased Mcl-1 level via inhibiting Akt signaling pathway in chemo-resistant cells. In addition, capivasertib not only decreased the viability of chemo-resistant esophageal cancer cells but also synergistically enhanced the effects of cisplatin. In multiple mouse models, representing both chemo-resistant and chemo-sensitive esophageal cancer, capivasertib administered at non-toxic doses demonstrated remarkable efficacy. It significantly extended the overall survival of the mice. Our research underscores the pivotal role of Akt-associated Mcl-1 upregulation in the development of chemo-resistance in esophageal cancer cells. Furthermore, it highlights the potential of capivasertib to reverse this resistance mechanism.
Collapse
Affiliation(s)
| | | | - Zhixiao Jiang
- Department of Thoracic Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 116 Zhuodaoquan South Load, Hongshan District, Wuhan, 430079, China
| | - Fei Xiong
- Department of Thoracic Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 116 Zhuodaoquan South Load, Hongshan District, Wuhan, 430079, China
| | - Sheng Wang
- Department of Thoracic Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 116 Zhuodaoquan South Load, Hongshan District, Wuhan, 430079, China
| |
Collapse
|
12
|
Raghuwanshi S, Zhang X, Arbieva Z, Khan I, Mohammed H, Wang Z, Domling A, Camacho CJ, Gartel AL. Novel FOXM1 inhibitor STL001 sensitizes human cancers to a broad-spectrum of cancer therapies. Cell Death Discov 2024; 10:211. [PMID: 38697979 PMCID: PMC11066125 DOI: 10.1038/s41420-024-01929-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/14/2024] [Accepted: 03/22/2024] [Indexed: 05/05/2024] Open
Abstract
Forkhead box protein M1 (FOXM1) is often overexpressed in human cancers and strongly associated with therapy resistance and less good patient survival. The chemotherapy options for patients with the most aggressive types of solid cancers remain very limited because of the acquired drug resistance, making the therapy less effective. NPM1 mutation through the inactivation of FOXM1 via FOXM1 relocalization to the cytoplasm confers more favorable treatment outcomes for AML patients, confirming FOXM1 as a crucial target to overcome drug resistance. Pharmacological inhibition of FOXM1 could be a promising approach to sensitize therapy-resistant cancers. Here, we explore a novel FOXM1 inhibitor STL001, a first-generation modification drug of our previously reported FOXM1 inhibitor STL427944. STL001 preserves the mode of action of the STL427944; however, STL001 is up to 50 times more efficient in reducing FOXM1 activity in a variety of solid cancers. The most conventional cancer therapies studied here induce FOXM1 overexpression in solid cancers. The therapy-induced FOXM1 overexpression may explain the failure or reduced efficacy of these drugs in cancer patients. Interestingly, STL001 increased the sensitivity of cancer cells to conventional cancer therapies by suppressing both the high-endogenous and drug-induced FOXM1. Notably, STL001 does not provide further sensitization to FOXM1-KD cancer cells, suggesting that the sensitization effect is conveyed specifically through FOXM1 suppression. RNA-seq and gene set enrichment studies revealed prominent suppression of FOXM1-dependent pathways and gene ontologies. Also, gene regulation by STL001 showed extensive overlap with FOXM1-KD, suggesting a high selectivity of STL001 toward the FOXM1 regulatory network. A completely new activity of FOXM1, mediated through steroid/cholesterol biosynthetic process and protein secretion in cancer cells was also detected. Collectively, STL001 offers intriguing translational opportunities as combination therapies targeting FOXM1 activity in a variety of human cancers driven by FOXM1.
Collapse
Affiliation(s)
| | - Xu Zhang
- University of Illinois at Chicago, Department of Medicine, Chicago, IL, USA
| | - Zarema Arbieva
- University of Illinois at Chicago, Department of Medicine, Chicago, IL, USA
| | - Irum Khan
- Northwestern University, Chicago, IL, USA
| | - Hisham Mohammed
- Oregon Health & Science University, Knight Cancer Institute, School of Medicine, Chicago, IL, USA
| | - Z Wang
- The Czech Advanced Technology and Research Institute (CATRIN) of Palacký University, Chicago, IL, USA
| | - Alexander Domling
- The Czech Advanced Technology and Research Institute (CATRIN) of Palacký University, Chicago, IL, USA.
| | - Carlos Jaime Camacho
- Department of Computational and Systems Biology, University of Pittsburgh, Chicago, IL, USA.
| | - Andrei L Gartel
- University of Illinois at Chicago, Department of Medicine, Chicago, IL, USA.
| |
Collapse
|
13
|
Yang R, Zhang H, Chen Z, Zhang T, Wei P, Liang H, He Y, Zheng C, Wang X, Zhang Y. Drug resistance‑related gene targets and molecular mechanisms in the A2780/Taxol‑resistant epithelial ovarian cancer cell line. Oncol Lett 2024; 27:232. [PMID: 38586210 PMCID: PMC10996034 DOI: 10.3892/ol.2024.14365] [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: 10/24/2023] [Accepted: 03/08/2024] [Indexed: 04/09/2024] Open
Abstract
Epithelial ovarian cancer (EOC) is a fatal gynecological malignant tumor with a low 5-year survival rate. The use of the first-line chemotherapeutic drug, paclitaxel, for the treatment of EOC is associated with resistance, often leading to treatment failure. The present study investigated the gene targets in an A2780 paclitaxel-resistant EOC cell line (A2780/Taxol), and the potential underlying mechanisms using transcriptome sequencing technology and bioinformatics analysis. The transcriptome of the A2780/Taxol cell line was sequenced, and 498 differentially expressed genes were obtained contained in the Gene Expression Omnibus dataset. Further bioinformatics analysis revealed that matrix metalloproteinase 1 (MMP1), zyxin (ZYX) and Unc-5 netrin receptor C (UNC5C) may be gene targets related to paclitaxel resistance. Moreover, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis indicated that a potential mechanism associated with paclitaxel resistance was related to cell migration. Furthermore, the expression levels of MMP1, ZYX and UNC5C were verified using western blotting, immunofluorescence and immunohistochemistry in vitro. The results revealed that the expression levels of MMP1 and ZYX were significantly increased in A2780/Taxol cells, while UNC5C expression was significantly decreased, which was consistent with the results of the transcriptome sequencing. The present study demonstrated that MMP1, ZYX and UNC5C may be the gene targets associated with paclitaxel resistance in EOC. These genes have potential to be used as molecular markers for EOC drug therapy, targeted elimination of drug resistance, and evaluation of treatment efficacy and patient prognosis.
Collapse
Affiliation(s)
- Ruihui Yang
- Department of Cell Biology and Medical Genetics, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Huainian Zhang
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zexin Chen
- Department of Cell Biology and Medical Genetics, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Tao Zhang
- Department of Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510080, P.R. China
| | - Peng Wei
- Department of Physiology, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Huaguo Liang
- Department of Cell Biology and Medical Genetics, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Yaoyao He
- Department of Cell Biology and Medical Genetics, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Changtao Zheng
- Department of Cell Biology and Medical Genetics, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Xicheng Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong 510080, P.R. China
| | - Yongli Zhang
- Department of Cell Biology and Medical Genetics, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| |
Collapse
|
14
|
Gartel A, Raghuwanshi S, Zhang X, Arbieva Z, Khan I, Wang Z, Domling A, Camacho C. [WITHDRAWN] Novel FOXM1 inhibitor STL001 sensitizes human cancers to a broad-spectrum of cancer therapies. RESEARCH SQUARE 2024:rs.3.rs-3711759. [PMID: 38234752 PMCID: PMC10793495 DOI: 10.21203/rs.3.rs-3711759/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
The full text of this preprint has been withdrawn by the authors while they make corrections to the work. Therefore, the authors do not wish this work to be cited as a reference. Questions should be directed to the corresponding author.
Collapse
|
15
|
[WITHDRAWN] Novel FOXM1 inhibitor STL001 sensitizes human cancers to a broad-spectrum of cancer therapies. RESEARCH SQUARE 2024:rs.3.rs-3711759. [PMID: 38234752 PMCID: PMC10793495 DOI: 10.21203/rs.3.rs-3711759/v2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
The full text of this preprint has been withdrawn by the authors while they make corrections to the work. Therefore, the authors do not wish this work to be cited as a reference. Questions should be directed to the corresponding author.
Collapse
|
16
|
Song K, Ma C, Gu B, Wang B, Ma H, Deng X, Chen H. Molecular mechanism underlying epithelial-mesenchymal transformation and cisplatin resistance in esophageal squamous cell carcinoma. Thorac Cancer 2023; 14:3069-3079. [PMID: 37718469 PMCID: PMC10626249 DOI: 10.1111/1759-7714.15094] [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: 07/23/2023] [Revised: 08/20/2023] [Accepted: 08/21/2023] [Indexed: 09/19/2023] Open
Abstract
Esophageal cancer (EC) occupies the seventh spot of the most prevalent malignancy cancer ailments worldwide and the sixth leading cause of cancer-related death. Esophageal squamous cell carcinoma (ESCC) is also the most predominant histological subtype of EC, and cisplatin (DDP) is commonly used as a first-line chemotherapeutic drug for the late advanced stages of the disease. However, the emergence of drug resistance during clinical treatment possesses a significant challenge to the therapeutic success and patient outcomes. Collectively, the epithelial-mesenchymal transformation (EMT) is a process in which transcription factors are induced to regulate the expression of epithelial and stromal markers to promote the differentiation of epithelial cells into stromal cells. Recent studies have demonstrated a close association between EMT and chemotherapy resistance in tumor cells, with concrete evidence of reciprocal reinforcement. Therefore, in this review, we elucidate the molecular mechanism underlying ESCC, shed light on the mechanisms driving DDP resistance, and provide insights into the intricate interplay between EMT and ESCC. We have aimed to provide some new hypotheses and perspectives that may address-inform future therapeutic strategies for ESCC treatment.
Collapse
Affiliation(s)
- Kewei Song
- The Second Clinical Medical College, Lanzhou UniversityLanzhouChina
- Department of Public HealthJining No.1 People's HospitalJiningChina
| | - Chenhui Ma
- The Second Clinical Medical College, Lanzhou UniversityLanzhouChina
| | - Baohong Gu
- The Second Clinical Medical College, Lanzhou UniversityLanzhouChina
| | - Bofang Wang
- The Second Clinical Medical College, Lanzhou UniversityLanzhouChina
| | - Huanhuan Ma
- The Second Clinical Medical College, Lanzhou UniversityLanzhouChina
| | - Xiaobo Deng
- The Second Clinical Medical College, Lanzhou UniversityLanzhouChina
| | - Hao Chen
- Department of Tumor SurgeryLanzhou University Second HospitalLanzhouChina
- Key Laboratory of Digestive System Tumors of Gansu ProvinceLanzhouChina
| |
Collapse
|
17
|
Liu WS, Wu LL, Chen CM, Zheng H, Gao J, Lu ZM, Li M. Lipid-hybrid cell-derived biomimetic functional materials: A state-of-the-art multifunctional weapon against tumors. Mater Today Bio 2023; 22:100751. [PMID: 37636983 PMCID: PMC10448342 DOI: 10.1016/j.mtbio.2023.100751] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023] Open
Abstract
Tumors are among the leading causes of death worldwide. Cell-derived biomimetic functional materials have shown great promise in the treatment of tumors. These materials are derived from cell membranes, extracellular vesicles and bacterial outer membrane vesicles and may evade immune recognition, improve drug targeting and activate antitumor immunity. However, their use is limited owing to their low drug-loading capacity and complex preparation methods. Liposomes are artificial bionic membranes that have high drug-loading capacity and can be prepared and modified easily. Although they can overcome the disadvantages of cell-derived biomimetic functional materials, they lack natural active targeting ability. Lipids can be hybridized with cell membranes, extracellular vesicles or bacterial outer membrane vesicles to form lipid-hybrid cell-derived biomimetic functional materials. These materials negate the disadvantages of both liposomes and cell-derived components and represent a promising delivery platform in the treatment of tumors. This review focuses on the design strategies, applications and mechanisms of action of lipid-hybrid cell-derived biomimetic functional materials and summarizes the prospects of their further development and the challenges associated with it.
Collapse
Affiliation(s)
- Wen-Shang Liu
- Department of Dermatology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, 200011, China
| | - Li-Li Wu
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Cui-Min Chen
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Hao Zheng
- Department of Gastrointestinal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Zheng-Mao Lu
- Department of Gastrointestinal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Meng Li
- Department of Dermatology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, 200011, China
| |
Collapse
|
18
|
Wang CD, Ma BQ, Yi BS. Resveratrol reverses drug resistance of esophageal cancer cell line Eca109/DDP via regulating ferroptosis. Shijie Huaren Xiaohua Zazhi 2023; 31:477-484. [DOI: 10.11569/wcjd.v31.i12.477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/28/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Resveratrol not only has anti-tumor effects, but also can enhance the chemosensitivity of tumor cells to a variety of chemotherapeutic agents. However, its effect on cisplatin sensitivity of drug-resistant esophageal cancer cells remains unclear.
AIM To investigate whether resveratrol reverses the drug resistance of esophageal cancer Eca109/DDP cells and to explore the potential mechanism involved from the perspective of ferroptosis.
METHODS The optimal treatment time and concentration of resveratrol and cisplatin were determined by MTT assay. Cell proliferation and the intracellular levels of malon-dialdehyde (MDA), glutathione (GSH), reactive oxygen species (ROS), and ferrous iron were detected. The protein expression of the ferroptosis-related molecules acyl coenzyme A synthetase long chain family member 4 (ACSL4), ferritin heavy chain (FTH), glutathione peroxidase 4 (GPX4), and tumor protein 53 (P53) was detected by Western blot assay.
RESULTS MTT assay showed that compared with cisplatin alone, resveratrol combined with cisplatin significantly inhibited the growth of Eca109/DDP cells (P < 0.05). The combination of resveratrol and cisplatin not only reduced the number of colonies formed (P < 0.05), but also increased the levels of ferrous iron, MDA, and ROS (P < 0.05) and decreased the level of GSH (P < 0.05) in Eca109/DDP cells. The inhibitory effect of resveratrol on Eca109/DDP cell proliferation was partially reversed by ferrostatin-1 (Fer-1) and deferoxamine (DFO) (P < 0.05). Western blot analysis showed that compared with other groups, the protein expression of FTH and GPX4 in the resveratrol and cisplatin combination group was significantly decreased (P < 0.05), and the protein expression of ACSL4 and P53 was significantly increased (P < 0.05).
CONCLUSION Resveratrol can inhibit cell proliferation and reverse cisplatin resistance by regulating ferroptosis in Eca109/DDP cells.
Collapse
Affiliation(s)
- Chen-Deng Wang
- Department of Gastrointestinal, Hernia, Bariatric metabolic, and Trauma Surgery, Lishui City People's Hospital, Lishui 323000, Zhejiang Province, China
| | - Bai-Qiang Ma
- Department of Gastrointestinal, Hernia, Bariatric metabolic, and Trauma Surgery, Lishui City People's Hospital, Lishui 323000, Zhejiang Province, China
| | - Bi-Shun Yi
- Department of Gastrointestinal, Hernia, Bariatric metabolic, and Trauma Surgery, Lishui City People's Hospital, Lishui 323000, Zhejiang Province, China
| |
Collapse
|
19
|
Reis JSD, Santos MARDC, da Costa KM, Freire-de-Lima CG, Morrot A, Previato JO, Previato LM, da Fonseca LM, Freire-de-Lima L. Increased Expression of the Pathological O-glycosylated Form of Oncofetal Fibronectin in the Multidrug Resistance Phenotype of Cancer Cells. Matrix Biol 2023; 118:47-68. [PMID: 36882122 DOI: 10.1016/j.matbio.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 02/15/2023] [Accepted: 03/03/2023] [Indexed: 03/07/2023]
Abstract
Changes in protein glycosylation are a hallmark of transformed cells and modulate numerous phenomena associated with cancer progression, such as the acquisition of multidrug resistance (MDR) phenotype. Different families of glycosyltransferases and their products have already been described as possible modulators of the MDR phenotype. Among the glycosyltransferases intensively studied in cancer research, UDP-N-acetyl-D-galactosamine:polypeptide N-acetylgalactosaminyltransferase-6 (pp-GalNAc-T6), which is widely expressed in many organs and tissues, stands out. Its influence in several events associated with kidney, oral, pancreatic, renal, lung, gastric and breast cancer progression has already been described. However, its participation in the MDR phenotype has never been studied. Here, we demonstrate that human breast adenocarcinoma MCF-7 MDR cell lines, generated by chronic exposure to doxorubicin, in addition to exhibiting increased expression of proteins belonging to the ABC superfamily (ABCC1 and ABCG2), and anti-apoptotic proteins (Blcl-2 and Bcl-xL), also present high expression of pp-GalNAc-T6, the enzyme currently proposed as the main responsible for the biosynthesis of oncofetal fibronectin (onf-FN), a major extracellular matrix component expressed by cancer cells and embryonic tissues, but absent in healthy cells. Our results show that onf-FN, which is generated by the addition of a GalNAc unit at a specific threonine residue inside the type III homology connective segment (IIICS) domain of FN, is strongly upregulated during the acquisition of the MDR phenotype. Also, the silencing of pp-GalNAc-T6, not only compromises the expression of the oncofetal glycoprotein, but also made the MDR cells more sensitive to all anticancer drugs tested, partially reversing the MDR phenotype. Taken together, our results demonstrate for the first time the upregulation of the O-glycosylated oncofetal fibronectin, as well as the direct participation of pp-GalNAc-T6 during the acquisition of a MDR phenotype in a breast cancer model, giving credence to the hypothesis that in transformed cells, glycosyltransferases and/or their products, such as unusual extracellular matrix glycoproteins can be used as potential therapeutic targets for the treatment of cancer.
Collapse
Affiliation(s)
- Jhenifer Santos Dos Reis
- Universidade Federal do Rio de Janeiro, Instituto de Biofísica Carlos Chagas Filho, Laboratório de Biologia Celular de Glicoconjugados, Rio de Janeiro, RJ 21941-902, Brazil
| | - Marcos André Rodrigues da Costa Santos
- Universidade Federal do Rio de Janeiro, Instituto de Biofísica Carlos Chagas Filho, Laboratório de Biologia Celular de Glicoconjugados, Rio de Janeiro, RJ 21941-902, Brazil
| | - Kelli Monteiro da Costa
- Universidade Federal do Rio de Janeiro, Instituto de Biofísica Carlos Chagas Filho, Laboratório de Biologia Celular de Glicoconjugados, Rio de Janeiro, RJ 21941-902, Brazil
| | - Celio Geraldo Freire-de-Lima
- Universidade Federal do Rio de Janeiro, Instituto de Biofísica Carlos Chagas Filho, Laboratório de Biologia Celular de Glicoconjugados, Rio de Janeiro, RJ 21941-902, Brazil
| | - Alexandre Morrot
- Universidade Federal do Rio de Janeiro, Faculdade de Medicina, Rio de Janeiro, RJ 21941-902, Brazil; Fiocruz, Instituto Oswaldo Cruz, Laboratório de Imunoparasitologia, Rio de Janeiro, RJ 21040-360, Brazil
| | - Jose Osvaldo Previato
- Universidade Federal do Rio de Janeiro, Instituto de Biofísica Carlos Chagas Filho, Laboratório de Biologia Celular de Glicoconjugados, Rio de Janeiro, RJ 21941-902, Brazil
| | - Lucia Mendonça Previato
- Universidade Federal do Rio de Janeiro, Instituto de Biofísica Carlos Chagas Filho, Laboratório de Biologia Celular de Glicoconjugados, Rio de Janeiro, RJ 21941-902, Brazil
| | - Leonardo Marques da Fonseca
- Universidade Federal do Rio de Janeiro, Instituto de Biofísica Carlos Chagas Filho, Laboratório de Biologia Celular de Glicoconjugados, Rio de Janeiro, RJ 21941-902, Brazil
| | - Leonardo Freire-de-Lima
- Universidade Federal do Rio de Janeiro, Instituto de Biofísica Carlos Chagas Filho, Laboratório de Biologia Celular de Glicoconjugados, Rio de Janeiro, RJ 21941-902, Brazil.
| |
Collapse
|
20
|
Guo X, Gao C, Yang DH, Li S. Exosomal circular RNAs: A chief culprit in cancer chemotherapy resistance. Drug Resist Updat 2023; 67:100937. [PMID: 36753923 DOI: 10.1016/j.drup.2023.100937] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/03/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Chemotherapy is one of the primary treatments for malignant tumors. However, the acquired drug resistance hinders clinical efficacy and leads to treatment failure in most patients. Exosomes are cell-derived vesicles with a diameter of 30-150 nm carrying and delivering substances such as DNAs, RNAs, lipids, and proteins for cellular communication in tumor development. Circular RNAs (circRNAs) present covalently closed-loop RNA structures, which regulate tumor cell proliferation, apoptosis, and metastasis by controlling different genes and signaling pathways. CircRNAs are abundant and stably expressed in exosomes. Recent studies have shown that they play critical roles in chemotherapy resistance in various cancers. In this review, we summarized the origin of exosomes and discussed the regulation mechanism of exosomal circRNAs in cancer drug resistance.
Collapse
Affiliation(s)
- Xu Guo
- Department of Neurosurgery, Cancer Hospital of Dalian University of Technology,Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province 110042, China
| | - Congying Gao
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Dong-Hua Yang
- New York College of Traditional Chinese Medicine, Mineola, NY, USA.
| | - Shenglong Li
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of Dalian University of Technology,Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang Liaoning Province 110042, China.
| |
Collapse
|
21
|
Zhang R, Dong M, Tu J, Li F, Deng Q, Xu J, He X, Ding J, Xia J, Sheng D, Chang Z, Ma W, Dong H, Zhang Y, Zhang L, Zhang L, Liu S. PMN-MDSCs modulated by CCL20 from cancer cells promoted breast cancer cell stemness through CXCL2-CXCR2 pathway. Signal Transduct Target Ther 2023; 8:97. [PMID: 36859354 PMCID: PMC9977784 DOI: 10.1038/s41392-023-01337-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 01/05/2023] [Accepted: 01/29/2023] [Indexed: 03/03/2023] Open
Abstract
Our previous studies have showed that C-C motif chemokine ligand 20 (CCL20) advanced tumor progression and enhanced the chemoresistance of cancer cells by positively regulating breast cancer stem cell (BCSC) self-renewal. However, it is unclear whether CCL20 affects breast cancer progression by remodeling the tumor microenvironment (TME). Here, we observed that polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) were remarkably enriched in TME of CCL20-overexpressing cancer cell orthotopic allograft tumors. Mechanistically, CCL20 activated the differentiation of granulocyte-monocyte progenitors (GMPs) via its receptor C-C motif chemokine receptor 6 (CCR6) leading to the PMN-MDSC expansion. PMN-MDSCs from CCL20-overexpressing cell orthotopic allograft tumors (CCL20-modulated PMN-MDSCs) secreted amounts of C-X-C motif chemokine ligand 2 (CXCL2) and increased ALDH+ BCSCs via activating CXCR2/NOTCH1/HEY1 signaling pathway. Furthermore, C-X-C motif chemokine receptor 2 (CXCR2) antagonist SB225002 enhanced the docetaxel (DTX) effects on tumor growth by decreasing BCSCs in CCL20high-expressing tumors. These findings elucidated how CCL20 modulated the TME to promote cancer development, indicating a new therapeutic strategy by interfering with the interaction between PMN-MDSCs and BCSCs in breast cancer, especially in CCL20high-expressing breast cancer.
Collapse
Affiliation(s)
- Rui Zhang
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai, 200032, China
| | - Mengxue Dong
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai, 200032, China
| | - Juchuanli Tu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai, 200032, China
| | - Fengkai Li
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai, 200032, China
| | - Qiaodan Deng
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai, 200032, China
| | - Jiahui Xu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai, 200032, China
| | - Xueyan He
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai, 200032, China
| | - Jiajun Ding
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai, 200032, China
- Breast Surgery, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Jie Xia
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai, 200032, China
| | - Dandan Sheng
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai, 200032, China
| | - Zhaoxia Chang
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai, 200032, China
| | - Wei Ma
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai, 200032, China
| | - Haonan Dong
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai, 200032, China
| | - Yi Zhang
- Department of Breast and Thyroid Surgery, Southwest Hospital, the First Affiliated Hospital of the Army Military Medical University, Chongqing, 400038, China
| | - Lixing Zhang
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai, 200032, China
| | - Lu Zhang
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai, 200032, China.
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; State Key Laboratory of Genetic Engineering; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; The Shanghai Key Laboratory of Medical Epigenetics; Shanghai Key Laboratory of Radiation Oncology; The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology; Shanghai Medical College; Fudan University, Shanghai, 200032, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
22
|
El-Tanani M, Al Khatib AO, Al-Najjar BO, Shakya AK, El-Tanani Y, Lee YF, Serrano-Aroca Á, Mishra V, Mishra Y, Aljabali AA, Goyal R, Negi P, Farani MR, Binabaj MM, Gholami A, Binabaj MM, Charbe NB, Tambuwala MM. Cellular and molecular basis of therapeutic approaches to breast cancer. Cell Signal 2023; 101:110492. [PMID: 36241056 DOI: 10.1016/j.cellsig.2022.110492] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/29/2022]
Abstract
In recent decades, there has been a significant amount of research into breast cancer, with some important breakthroughs in the treatment of both primary and metastatic breast cancers. It's a well-known fact that treating breast cancer is still a challenging endeavour even though physicians have a fantastic toolset of the latest treatment options at their disposal. Due to limitations of current clinical treatment options, traditional chemotherapeutic drugs, and surgical options are still required to address this condition. In recent years, there have been several developments resulting in a wide range of treatment options. This review article discusses the cellular and molecular foundation of chemotherapeutic drugs, endocrine system-based treatments, biological therapies, gene therapy, and innovative techniques for treating breast cancer.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan; Centre for Cancer Research and Cell Biology, Queen's University Belfast, Grosvenor Road, Belfast BT12 6BJ, Northern Ireland, UK; Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, UK.
| | - Arwa Omar Al Khatib
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan
| | - Belal O Al-Najjar
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan
| | - Ashok K Shakya
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan
| | - Yahia El-Tanani
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Grosvenor Road, Belfast BT12 6BJ, Northern Ireland, UK; Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford, UK
| | - Yin-Fai Lee
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, UK; School of Life Sciences, Faculty of Science and Engineering, Anglia Ruskin University, Cambridge CB1 1PT, UK; Neuroscience, Psychology & Behaviour, College of Life Sciences, University of Leicester, Leicester LE1 9HN, UK
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Laboratory, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Yachana Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Alaa A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid 566, Jordan
| | - Rohit Goyal
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan 173229, India
| | - Poonam Negi
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan 173229, India
| | - Marzieh Ramezani Farani
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), the Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), 1417614411 Tehran, Iran.
| | - Maryam Moradi Binabaj
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Amir Gholami
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Maryam Moradi Binabaj
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Nitin B Charbe
- Center for pharmacometrics and system pharmacology, department of pharmaceutics, college of pharmacy, University of Florida, FL, USA
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, UK; Neuroscience, Psychology & Behaviour, College of Life Sciences, University of Leicester, Leicester LE1 9HN, UK.
| |
Collapse
|
23
|
Harada K, Sakamoto N. Cancer organoid applications to investigate chemotherapy resistance. Front Mol Biosci 2022; 9:1067207. [PMID: 36582205 PMCID: PMC9792487 DOI: 10.3389/fmolb.2022.1067207] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022] Open
Abstract
In clinical practice, a large proportion of cancer patients receive chemotherapy, yet tumors persist or acquire resistance; removing this obstacle could help to lower the number of cancer-related fatalities. All areas of cancer research are increasingly using organoid technology, a culture technique that simulates the in vivo environment in vitro, especially in the quickly developing fields of anticancer drug resistance, drug-tolerant persisters, and drug screening. This review provides an overview of organoid technology, the use of organoids in the field of anticancer drug resistance research, their relevance to clinical information and clinical trials, and approaches to automation and high throughput.
Collapse
Affiliation(s)
- Kenji Harada
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan,Division of Pathology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Naoya Sakamoto
- Division of Pathology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan,*Correspondence: Naoya Sakamoto,
| |
Collapse
|
24
|
Liao W, Li Y, Wang J, Zhao M, Chen N, Zheng Q, Wan L, Mou Y, Tang J, Wang Z. Natural Products-Based Nanoformulations: A New Approach Targeting CSCs to Cancer Therapy. Int J Nanomedicine 2022; 17:4163-4193. [PMID: 36134202 PMCID: PMC9482958 DOI: 10.2147/ijn.s380697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/25/2022] [Indexed: 11/25/2022] Open
Abstract
Cancer stem cells (CSCs) lead to the occurrence and progression of cancer due to their strong tumorigenic, self-renewal, and multidirectional differentiation abilities. Existing cancer treatment methods cannot effectively kill or inhibit CSCs but instead enrich them and produce stronger proliferation, invasion, and metastasis capabilities, resulting in cancer recurrence and treatment resistance, which has become a difficult problem in clinical treatment. Therefore, targeting CSCs may be the most promising approach for comprehensive cancer therapy in the future. A variety of natural products (NP) have significant antitumor effects and have been identified to target and inhibit CSCs. However, pharmacokinetic defects and off-target effects have greatly hindered their clinical translation. NP-based nanoformulations (NPNs) have tremendous potential to overcome the disadvantages of NP against CSCs through site-specific delivery and by improving their pharmacokinetic parameters. In this review, we summarize the recent progress of NPNs targeting CSCs in cancer therapy, looking forward to transforming preclinical research results into clinical applications and bringing new prospects for cancer treatment.
Collapse
Affiliation(s)
- Wenhao Liao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Yuchen Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China.,College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Jing Wang
- Department of Obstetrics and Gynecology, Bishan Hospital of Traditional Chinese Medicine, Chongqing, People's Republic of China
| | - Maoyuan Zhao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Nianzhi Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Qiao Zheng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Lina Wan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Yu Mou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Jianyuan Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China.,TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Zhilei Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China.,TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| |
Collapse
|
25
|
Zhang R, Li S, Lan J, Li C, Du X, Dong W, Yu Q, Wang D. CNTN-1 Upregulation Induced by Low-Dose Cisplatin Promotes Malignant Progression of Lung Adenocarcinoma Cells via Activation of Epithelial-Mesenchymal Transition. Front Genet 2022; 13:891665. [PMID: 35711928 PMCID: PMC9196332 DOI: 10.3389/fgene.2022.891665] [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: 03/08/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Tumor metastasis and invasion are the main impediments to lung adenocarcinoma successful treatment. Previous studies demonstrate that chemotherapeutic agents can elevate the malignancy of cancer cells other than their therapeutic effects. In this study, the effects of transient low-dose cisplatin treatment on the malignant development of lung adenocarcinoma cells (A549) were detected, and the underlying epigenetic mechanisms were investigated. The findings showed that A549 cells exhibited epithelial-mesenchymal transition (EMT)-like phenotype along with malignant progression under the transient low-dose cisplatin treatment. Meanwhile, low-dose cisplatin was found to induce contactin-1 (CNTN-1) upregulation in A549 cells. Subsequently, we found that further overexpressing CNTN-1 in A549 cells obviously activated the EMT process in vitro and in vivo, and caused malignant development of A549 cells in vitro. Taken together, we conclude that low-dose cisplatin can activate the EMT process and resulting malignant progression through upregulating CNTN-1 in A549 cells. The findings provided new evidence that a low concentration of chemotherapeutic agents could facilitate the malignancy of carcinoma cells via activating the EMT process other than their therapeutic effects.
Collapse
Affiliation(s)
- Ruijie Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shengjin Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian Lan
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Changyi Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xianzhi Du
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weijie Dong
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qian Yu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Daoxin Wang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
26
|
Tang J, He J, Feng C, Tu C. Exosomal MiRNAs in Osteosarcoma: Biogenesis and Biological Functions. Front Pharmacol 2022; 13:902049. [PMID: 35592419 PMCID: PMC9110813 DOI: 10.3389/fphar.2022.902049] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/19/2022] [Indexed: 12/05/2022] Open
Abstract
MiRNAs are a group of non-coding RNA molecules that function in mRNA translational inhibition via base-pairing with complementary sequences in target mRNA. In oncology, miRNAs have raised great attention due to their aberrant expression and pivotal roles in the pathogenesis of multiple malignancies including osteosarcoma. MiRNAs can be transported by exosome, the nano-extracellular vesicle with a diameter of 30–150 nm. Recently, a growing number of studies have demonstrated that exosomal miRNAs play a critical role in tumor initiation and progression, by exerting multiple biological functions including metastasis, angiogenesis, drug resistance and immunosuppression. In this review, we aim to depict the biogenesis of exosomal miRNAs and summarize the potential diagnostic and therapeutic functions of exosomal miRNAs in osteosarcoma.
Collapse
Affiliation(s)
- Jinxin Tang
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Xiangya School of Medicine, Central South University, Changsha, China
| | - Jieyu He
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chengyao Feng
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|