1
|
Cai H, Tian S, Liu A, Xie G, Zhang H, Wu X, Wan J, Li S. Relationship between CTF1 gene expression and prognosis and tumor immune microenvironment in glioma. Eur J Med Res 2025; 30:17. [PMID: 39780198 PMCID: PMC11715937 DOI: 10.1186/s40001-024-02192-w] [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: 05/01/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE This study aimed to evaluate CTF1 expression in glioma, its relationship to patient prognosis and the tumor immune microenvironment, and effects on glioma phenotypes to identify a new therapeutic target for treating glioma precisely. METHODS We initially assessed the expression of CTF1, a member of the IL-6 family, in glioma, using bioinformatics tools and publicly available databases. Furthermore, we examined the correlation between CTF1 expression and tumor prognosis, DNA methylation patterns, m6A-related genes, potential biological functions, the immune microenvironment, and genes associated with immune checkpoints. We also explored potential associations with drug sensitivity. To assess the impact on glioma cell proliferation and apoptosis, we employed various assays, including the Cell Counting Kit-8, colony formation assay, and flow cytometry. RESULTS CTF1 gene and protein expression were significantly elevated in glioma tissues, and correlated with malignancy and poor prognosis. CTF1 was an independent prognostic factor and negatively associated with DNA methylation. The involvement of CTF1 in m6A modifications contributed to glioma progression. Enrichment analysis revealed immune response pathways linked with CTF1 in glioma, including natural killer cell cytotoxicity, NOD-like receptor signaling, Toll-like receptor signaling, antigen processing, chemokine signaling, and cytokine receptor interactions. CTF1 expression correlated positively with pathways related to apoptosis, inflammation, proliferation, and epithelial-mesenchymal transition, and PI3K-AKT-mTOR signaling. Additionally, CTF1 expression was positively associated with macrophage, eosinophil, and neutrophil contents and immune checkpoint-related genes, but negatively associated with sensitivity to 14 drugs. In vitro experiments confirmed that CTF1 knockdown inhibited glioma cell proliferation and promoted apoptosis. CONCLUSION This study identifies CTF1 as a significant independent prognostic factor that is closely associated with the tumor immune microenvironment in glioma. Additionally, reduced expression of CTF1 suppresses the proliferation and induces apoptosis of glioma cells in vitro. Consequently, CTF1 is a potentially promising novel therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Hongqing Cai
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, People's Republic of China
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shen Tian
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, People's Republic of China
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Angsi Liu
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, People's Republic of China
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guanchao Xie
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, People's Republic of China
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, No 678 Furong Road, Economic and Technological Development Zone, Hefei, 230000, Anhui, People's Republic of China
| | - Hongsheng Zhang
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, People's Republic of China
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, No 678 Furong Road, Economic and Technological Development Zone, Hefei, 230000, Anhui, People's Republic of China
| | - Xiaogang Wu
- Department of Neurosurgery, No. 901 Hospital of the Chinese People's Liberation Army Logistic Support Force, No 424 Changjiang West Road, Shushan District, Hefei, Anhui, 230000, People's Republic of China.
| | - Jinghai Wan
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, People's Republic of China.
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, No 678 Furong Road, Economic and Technological Development Zone, Hefei, 230000, Anhui, People's Republic of China.
| | - Sai Li
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, People's Republic of China.
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, No 678 Furong Road, Economic and Technological Development Zone, Hefei, 230000, Anhui, People's Republic of China.
| |
Collapse
|
2
|
Feng QS, Shan XF, Yau V, Cai ZG, Xie S. Facilitation of Tumor Stroma-Targeted Therapy: Model Difficulty and Co-Culture Organoid Method. Pharmaceuticals (Basel) 2025; 18:62. [PMID: 39861125 PMCID: PMC11769033 DOI: 10.3390/ph18010062] [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: 12/10/2024] [Revised: 12/28/2024] [Accepted: 01/05/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Tumors, as intricate ecosystems, comprise oncocytes and the highly dynamic tumor stroma. Tumor stroma, representing the non-cancerous and non-cellular composition of the tumor microenvironment (TME), plays a crucial role in oncogenesis and progression, through its interactions with biological, chemical, and mechanical signals. This review aims to analyze the challenges of stroma mimicry models, and highlight advanced personalized co-culture approaches for recapitulating tumor stroma using patient-derived tumor organoids (PDTOs). Methods: This review synthesizes findings from recent studies on tumor stroma composition, stromal remodeling, and the spatiotemporal heterogeneities of the TME. It explores popular stroma-related models, co-culture systems integrating PDTOs with stromal elements, and advanced techniques to improve stroma mimicry. Results: Stroma remodeling, driven by stromal cells, highlights the dynamism and heterogeneity of the TME. PDTOs, derived from tumor tissues or cancer-specific stem cells, accurately mimic the tissue-specific and genetic features of primary tumors, making them valuable for drug screening. Co-culture models combining PDTOs with stromal elements effectively recreate the dynamic TME, showing promise in personalized anti-cancer therapy. Advanced co-culture techniques and flexible combinations enhance the precision of tumor-stroma recapitulation. Conclusions: PDTO-based co-culture systems offer a promising platform for stroma mimicry and personalized anti-cancer therapy development. This review underscores the importance of refining these models to advance precision medicine and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Qiu-Shi Feng
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22# Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (Q.-S.F.); (X.-F.S.)
| | - Xiao-Feng Shan
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22# Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (Q.-S.F.); (X.-F.S.)
| | - Vicky Yau
- Division of Oral and Maxillofacial Surgery, Columbia Irving Medical Center, New York City, NY 10027, USA;
| | - Zhi-Gang Cai
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22# Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (Q.-S.F.); (X.-F.S.)
| | - Shang Xie
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22# Zhongguancun South Avenue, Haidian District, Beijing 100081, China; (Q.-S.F.); (X.-F.S.)
| |
Collapse
|
3
|
Xu C, Zhang C, Ganesan K, Qui C, Tang H, Gao F, Liu Q, Wu J, Sui Y, Li P, Zhang J, Chen J. Anti-migratory Properties of Cryoprotective Isoliquiritigenin-zein Phosphatidylcholine Nanoparticles Prevent Triple-negative Breast Cancer through PI3K-mTOR and MMP2/9 Pathways. Curr Med Chem 2025; 32:1770-1788. [PMID: 37936460 DOI: 10.2174/0109298673259973231023110945] [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: 05/13/2023] [Revised: 06/26/2023] [Accepted: 09/15/2023] [Indexed: 11/09/2023]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC), an aggressive type of breast cancer, remains difficult to treat. Isoliquiritigenin (ISL) is a bioactive compound that is insoluble in water and exhibits significant anti-TNBC activity. METHODS We previously prepared oral aqueous ISL@ZLH NPs; however, they were less stable in a freezing environment. Hence, the present study aimed to improve the stability of ISL@ZLH NPs using cryoprotectants that can withstand long storage times and are effective in TNBC treatment by creating an efficient oral drug delivery system. Freeze-dried ISL@ZLH NP powder was prepared by solvent evaporation, followed by the addition of trehalose and sucrose. The freeze-dried ISL@ZLH NP pow was optimized and characterized. The anti-TNBC efficacy and pharmacokinetics of the ISL@ZLH NP-pow were examined in plasma and organs, compared with those of aqueous ISL@ZLH NPs. RESULT The ideal particle size of the ISL@ZLH NP pow was 118 nm, which was not filtered out by the glomerulus and allowed the drug to be delivered to the lesions more effectively. Cellular uptake and biodistribution of the ISL@ZLH NP-pow in vivo and in vitro showed prolonged storage in the organs. In addition, cryopreserved ISL@ZLH NP-treated tumors showed significant anti- proliferative and anti-migratory effects through the downregulation of the PI3K-Akt-mToR and MMP2/9 signaling pathways. CONCLUSION These results suggest that oral ingestion of cryopreserved ISL@ZLH NP has the potential for long-term storage and can be employed as a clinical therapeutic approach to treat TNBC.
Collapse
Affiliation(s)
- Cong Xu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Cheng Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kumar Ganesan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chen Qui
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Department of Breast Oncology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Fei Gao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qingqing Liu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jianming Wu
- State Key Laboratory of Oncology in South China, Department of Breast Oncology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Yue Sui
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Peng Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Jinming Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianping Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
4
|
He J, Guo J, Sun P. Prognostic value of CTF1 in glioma and its role in the tumor microenvironment. Transl Cancer Res 2024; 13:6862-6879. [PMID: 39816535 PMCID: PMC11730202 DOI: 10.21037/tcr-24-1258] [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: 07/21/2024] [Accepted: 11/10/2024] [Indexed: 01/18/2025]
Abstract
Background Within the realm of primary brain tumors, specifically glioblastoma (GBM), presents a notable obstacle due to their unfavorable prognosis and differing median survival rates contingent upon tumor grade and subtype. Despite a plethora of research connecting cardiotrophin-1 (CTF1) modifications to a range of illnesses, its correlation with glioma remains uncertain. This study investigated the clinical value of CTF1 in glioma and its potential as a biomarker of the disease. Methods Glioma project in The Cancer Genome Atlas (TCGA) database served as the training cohort, and CGGA 325 series in the Chinese Glioma Genome Atlas (CGGA) database served as the external independent validation cohort. First, the difference in the expression level of CTF1 between glioma tissue and normal tissue was analyzed, and the results were verified with the CGGA database. The relationship between CTF1 expression and the prognosis of glioma patients was evaluated using Univariate and Multivariate Cox analysis and the Kaplan-Meier (KM) curve. We used CIBERSOFT to explore the association between CTF1 and immune cell infiltration in GBM, as well as performing gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) analyses. Furthermore, we analyzed the relationship between CTF1 and gene mutations and drug sensitivity. Using Weighted gene co-expression network analysis (WGCNA) analysis, we pinpointed the gene set most correlated with CTF1 and conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) gene enrichment analyses to anticipate the pathways that could be influenced by CTF1. Finally, we constructed a nomogram using a multifactorial regression model to further predict patient prognosis. Results CTF1 expression was significantly elevated in glioma tissues compared to normal tissues in the TCGA dataset (P<0.001) and was associated with poorer survival in both TCGA and CGGA datasets (P<0.001). Receiver operating characteristic (ROC) analysis demonstrated the diagnostic potential of CTF1, with an area under the curve (AUC) of 0.889 [95% confidence interval (CI): 0.803-0.974] in TCGA and 0.664 (95% CI: 0.599-0.729) in CGGA. High CTF1 levels were correlated with advanced glioma grades, and Cox regression analysis identified CTF1 as an independent risk factor. A nomogram incorporating CTF1 levels, isocitrate dehydrogenase 1 (IDH1) mutation status, O6-methylguanine-DNA methyltransferase (MGMT) methylation status, age, and gender were developed and validated to predict 1- and 2-year survival probabilities. In GBM, drug sensitivity analysis revealed significant associations between CTF1 expression and responsiveness to gemcitabine, dasatinib, and other agents. CTF1 expression was also linked to immune infiltration (monocytes, neutrophils, M0 macrophages) and pathways involved in tumor progression, including IL2_STAT5, P53, and IL6_JAK_STAT3 signaling pathways. Conclusions CTF1 could serve as a prognostic marker for glioma. It acts as a predictive indicator and is associated with immune cell infiltration in GBM. These findings provide a foundation for further research into the molecular function of CTF1 and offer new insights for exploring the underlying mechanisms and developing treatments for glioma.
Collapse
Affiliation(s)
- Jiakai He
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Radiation Oncology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Jiayin Guo
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Pengfei Sun
- Department of Radiation Oncology, The Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
5
|
Pérez-Mato M, Dopico-López A, Akkoc Y, López-Amoedo S, Correa-Paz C, Candamo-Lourido M, Iglesias-Rey R, López-Arias E, Bugallo-Casal A, da Silva-Candal A, Bravo SB, Chantada-Vázquez MDP, Arias S, Santamaría-Cadavid M, Estany-Gestal A, Zaghmi A, Gauthier MA, Gutiérrez-Fernández M, Martin A, Llop J, Rodríguez C, Almeida Á, Migliavacca M, Polo E, Pelaz B, Gozuacik D, El Yamani N, SenGupta T, Rundén-Pran E, Vivancos J, Castellanos M, Díez-Tejedor E, Sobrino T, Rabinkov A, Mirelman D, Castillo J, Campos F. Preclinical validation of human recombinant glutamate-oxaloacetate transaminase for the treatment of acute ischemic stroke. iScience 2024; 27:111108. [PMID: 39524351 PMCID: PMC11543921 DOI: 10.1016/j.isci.2024.111108] [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: 04/11/2024] [Revised: 08/21/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
The blood enzyme glutamate-oxaloacetate transaminase (GOT) has been postulated as an effective therapeutic to protect the brain during stroke. To demonstrate its potential clinical utility, a new human recombinant form of GOT (rGOT) was produced for medical use. We tested the pharmacokinetics and evaluated the protective efficacy of rGOT in rodent and non-human primate models that reflected clinical stroke conditions. We found that continuous intravenous administration of rGOT within the first 8 h after ischemic onset significantly reduced the infarct size in both severe (30%) and mild lesions (48%). Cerebrospinal fluid and proteomics analysis, in combination with positron emission tomography imaging, indicated that rGOT can reach the brain and induce cytoprotective autophagy and induce local protection by alleviating neuronal apoptosis. Our results suggest that rGOT can be safely used immediately in patients suspected of having a stroke. This study requires further validation in clinical stroke populations.
Collapse
Affiliation(s)
- María Pérez-Mato
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research – IdiPAZ (La Paz University Hospital- Universidad Autónoma de Madrid), 28029 Madrid, Spain
| | - Antonio Dopico-López
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Yunus Akkoc
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul 34450, Turkey
- Department of Medical Biology, Koç University School of Medicine, Istanbul 34450, Turkey
| | - Sonia López-Amoedo
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Clara Correa-Paz
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - María Candamo-Lourido
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Ramón Iglesias-Rey
- Neuroimaging and Biotechnology Laboratory Group (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Esteban López-Arias
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Ana Bugallo-Casal
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Andrés da Silva-Candal
- Neurology Service, University Hospital Complex of A Coruña, A Coruña Biomedical Research Institute, 15006 A Coruña, Spain
| | - Susana B. Bravo
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - María del Pilar Chantada-Vázquez
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Research Unit, Lucus Augusti University Hospital (HULA), Servizo Galego de Saúde (SERGAS), 27002 Lugo, Spain
| | - Susana Arias
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, 15706 Santiago de Compostela, Spain
| | - María Santamaría-Cadavid
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, 15706 Santiago de Compostela, Spain
| | - Ana Estany-Gestal
- Unit of Methodology of the Research, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Ahlem Zaghmi
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC J3X 1S2, Canada
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Marc A. Gauthier
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC J3X 1S2, Canada
| | - María Gutiérrez-Fernández
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research – IdiPAZ (La Paz University Hospital- Universidad Autónoma de Madrid), 28029 Madrid, Spain
| | - Abraham Martin
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain
- Ikerbasque Basque Foundation for Science, 48009 Bilbao, Spain
| | - Jordi Llop
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 San Sebastian, Spain
| | - Cristina Rodríguez
- Institute of Functional Biology and Genomics (IBFG), CSIC, University of Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, CSIC, University of Salamanca, 37007 Salamanca, Spain
| | - Ángeles Almeida
- Institute of Functional Biology and Genomics (IBFG), CSIC, University of Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, CSIC, University of Salamanca, 37007 Salamanca, Spain
| | - Martina Migliavacca
- Center for Research in Biological Chemistry and Molecular Materials (CiQUS), University of Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Ester Polo
- Center for Research in Biological Chemistry and Molecular Materials (CiQUS), University of Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Beatriz Pelaz
- Center for Research in Biological Chemistry and Molecular Materials (CiQUS), University of Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Devrim Gozuacik
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul 34450, Turkey
- Department of Medical Biology, Koç University School of Medicine, Istanbul 34450, Turkey
| | - Naouale El Yamani
- Health Effects Laboratory, Department for Environmental Chemistry, NILU-Norwegian Institute for Air Research, 2027 Kjeller, Norway
| | - Tanima SenGupta
- Health Effects Laboratory, Department for Environmental Chemistry, NILU-Norwegian Institute for Air Research, 2027 Kjeller, Norway
| | - Elise Rundén-Pran
- Health Effects Laboratory, Department for Environmental Chemistry, NILU-Norwegian Institute for Air Research, 2027 Kjeller, Norway
| | - José Vivancos
- Stroke Unit, Department of Neurology, Hospital Universitario de La Princesa & Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain
| | - Mar Castellanos
- Neurology Service, University Hospital Complex of A Coruña, A Coruña Biomedical Research Institute, 15006 A Coruña, Spain
| | - Exuperio Díez-Tejedor
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research – IdiPAZ (La Paz University Hospital- Universidad Autónoma de Madrid), 28029 Madrid, Spain
| | - Tomás Sobrino
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Aharon Rabinkov
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - David Mirelman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory Group (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Francisco Campos
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| |
Collapse
|
6
|
Tan T, Chang W, Wang TL, Chen W, Chen X, Yang C, Yang D. pH-Responsive Charge-Reversal Smart Nanoparticles for Co-Delivery of Mitoxantrone and Copper Ions to Enhance Breast Cancer Chemo-Chemodynamic Combination Therapy. Int J Nanomedicine 2024; 19:11445-11462. [PMID: 39530107 PMCID: PMC11552415 DOI: 10.2147/ijn.s479125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Purpose The poor delivery and limited penetration of nanoparticles into breast cancer tumors remain essential challenges for effective anticancer therapy. This study aimed to design a promising nanoplatform with efficient tumor targeting and penetration capability for effective breast cancer therapy. Methods A pH-sensitive mitoxantrone (MTO) and copper ion-loaded nanosystem functionalized with cyclic CRGDfK and r9 peptide (TPRN-CM) was rationally designed for chemo-chemodynamic combination therapy. TPRN-CM would be quiescent in blood circulation with the CRGDfK peptide on the surface of the nanoparticle to improve its targeting to the tumor. Then, the structure of TPRN-CM changes in the acidic tumor microenvironment, and the r9 peptide can be exposed to make a surface charge reversal to promote deep penetration in the tumor and facilitate their internalization by cancer cells, which was characterized using transmission electron microscopy, dynamic light scattering, flame atomic absorption, etc. The drug release behavior, anti-tumor effects in vivo and in vitro, and the biosafety of the nanoplatform were evaluated. Results TPRN-CM exhibited remarkable capability to load MTO and Cu2+ with good stability in serum. It can achieve pH-responsive charge reversal, MTO, and Cu2+ release, and can further generate toxic hydroxyl radicals in the presence of glutathione (GSH) and H2O2. In vitro experiments demonstrated that this nanoplatform significantly inhibited proliferation, migration, invasion activities and 3D-tumorsphere growth. In vivo experiments suggested that rationally designed TPRN-CM can be effectively delivered to breast cancer tumors with deep tumor penetration, thereby resulting in a notable reduction in tumor growth and suppression of lung metastasis without causing any apparent side effects. Conclusion The constructed TPRN-CM nanoplatform integrated tumor targeting, tumor penetration, drug-responsive release, and chemo-chemodynamic combination therapy, thereby providing an intelligent drug delivery strategy to improve the efficacy of breast cancer treatment.
Collapse
Affiliation(s)
- Tao Tan
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, 519041, People’s Republic of China
| | - Weiyi Chang
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, 519041, People’s Republic of China
- College of Life Science, Jilin University, Changchun, 130012, People’s Republic of China
| | - Tian Long Wang
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, 519041, People’s Republic of China
| | - Wei Chen
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, 519041, People’s Republic of China
| | - Xiaobing Chen
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, 519041, People’s Republic of China
| | - Chunmiao Yang
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, 519041, People’s Republic of China
| | - Dongsheng Yang
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, 519041, People’s Republic of China
| |
Collapse
|
7
|
Gao J, Yu G, Yan Y, Hu W, Hu D, Wang W, Yang G, Wei J, Yang S. ITIH1 suppresses carcinogenesis in renal cell carcinoma through regulation of the NF‑κB signaling pathway. Exp Ther Med 2024; 28:368. [PMID: 39091412 PMCID: PMC11292172 DOI: 10.3892/etm.2024.12657] [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: 09/08/2023] [Accepted: 03/22/2024] [Indexed: 08/04/2024] Open
Abstract
Renal cell carcinoma (RCC) is a common malignancy of the urinary system. Although traditional therapies, such as surgery assisted with chemotherapy have improved the quality of life and survival time of patients with RCC, patients with metastasis or recurrence benefit little from such therapies. At present, little is known about the underlying mechanisms of RCC, rendering treatment selection and implementation challenging. Therefore, investigating the cause and underlying mechanisms of RCC remain of importance to explore potential new avenues for its treatment. Inter-α-trypsin inhibitor heavy chain 1 (ITIH1) is an inflammation-associated gene reported to suppress the progression of liver cancer. However, its role in RCC remains poorly understood. Therefore, the present study aimed to investigate the role and mechanism of ITIH1 in RCC. Based on data obtained from The Cancer Genome Atlas database, ITIH1 expression was demonstrated to be significantly higher in tumor tissues compared with normal tissues, which was in turn negatively associated with the survival of patients with RCC. However, in RCC cells, ITIH1 was shown to be expressed at significantly lower levels compared with those in HK-2 cells. The discrepancy between tissues and cell lines might be due to the different environment of cell growth. ITIH1 knockdown in RCC cells significantly increased cell proliferation and invasion whilst significantly decreasing the apoptosis rate, compared with those in control cells (without ITIH1 knockdown). By contrast, overexpression of ITIH1 significantly inhibited cell proliferation and invasion in RCC cells. In terms of western blotting results, the phosphorylation levels of NF-κB were significantly increased following ITIH1 knockdown. The protein expression level of IκB significantly decreased whereas that of IKK, Cyclin D1, proliferating cell nuclear antigen and α-smooth muscle actin were significantly increased in ITIH1-knockdown cells, compared with those in the control cells (without ITIH1 knockdown). This suggests that the NF-κB pathway may be activated after ITIH1 knockdown. Following treatment with the NF-κB pathway inhibitor JSH-23 in combination with ITIH1 knockdown, RCC cell proliferation and invasion were significantly reduced compared with those after ITIH1 knockdown alone. In summary, results from the present study suggest that ITIH1 can serve an inhibitory role in the progression of RCC, which could potentially be inhibited through the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Jing Gao
- Department of General Practice, Xujiahui Community Healthcare Center of Xuhui District of Shanghai, Shanghai 200030, P.R. China
| | - Gang Yu
- Department of Nephrology, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai 200233, P.R. China
| | - Yan Yan
- Department of General Practice, Xujiahui Community Healthcare Center of Xuhui District of Shanghai, Shanghai 200030, P.R. China
| | - Weifeng Hu
- Department of Nephrology, Naval Medical Center of People's Liberation Army, Shanghai 200052, P.R. China
| | - Dayong Hu
- Department of Nephrology, The Tenth People's Hospital of Tongji University, Shanghai 200072, P.R. China
| | - Weibing Wang
- Department of Epidemiology, School of Public Health of Fudan University, Shanghai 200032, P.R. China
| | - Guoxian Yang
- Department of General Practice, Xujiahui Community Healthcare Center of Xuhui District of Shanghai, Shanghai 200030, P.R. China
| | - Jing Wei
- Department of General Practice, Xujiahui Community Healthcare Center of Xuhui District of Shanghai, Shanghai 200030, P.R. China
| | - Shiquan Yang
- Department of General Practice, Xujiahui Community Healthcare Center of Xuhui District of Shanghai, Shanghai 200030, P.R. China
| |
Collapse
|
8
|
Strzelczyk J, Janas K, Strzelczyk JK, Chełmecka E, Kajdaniuk D, Kos-Kudła B. Evaluation of selected circulating cytokines from the IL-6 family - interleukin 6, oncostatin M, and cardiotrophin-1 - in gastro-entero-pancreatic and bronchial neuroendocrine tumours. Contemp Oncol (Pozn) 2024; 28:114-120. [PMID: 39421708 PMCID: PMC11480914 DOI: 10.5114/wo.2024.142584] [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: 07/17/2024] [Accepted: 08/08/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction The incidence of neuroendocrine tumours (NETs) increased over the last years. Most of them are non-functioning, and the course of the disease is asymptomatic for a long time. This results in late diagnosis at an advanced stage. The aim of our study was the evaluation of selected circulating cytokines of interleukin-6 family - interleukin 6 (IL-6), oncostatin M (OSM), and cardiotrophin-1 (CT1) - in NETs. Material and methods The study group comprised 80 patients (56%) in several subgroups, including gastroenteropancreatic (GEPNETs, n = 64, 80%) and bronchopulmonary neuroendocrine tumours (BPNETs, n = 16; 20%). Serum IL-6, OSM, and CT1 concentrations were tested using ELISA. Results The median concentration of IL-6 was 41.5 pg/ml in the study group and 32.6 pg/ml in the control group, and the difference was statistically significant (p < 0.001). The concentration of OSM was significantly lower in the study group than in the control group (p < 0.001), at 105.6 pg/ml and 115.5 pg/ml, respectively. There was a significant difference (p < 0.01) in concentration of CT1 in the study group (222.0 pg/ml) and controls (267.2 pg/ml). Our investigation into selected IL-6 family cytokines revealed differential modulation of signal transduction pathways. Conclusions These findings suggest that despite utilising a common signalling transducer, individual IL-6 family cytokines exert distinct biological effects on neuroendocrine tumour development. Notably, IL-6 appears to promote tumourigenesis, while OSM and CT1 exhibit inhibitory effects on gastro-entero-pancreatic and bronchial neuroendocrine tumour development. Further studies are necessary to validate the diagnostic utility of IL-6 family cytokines in NETs.
Collapse
Affiliation(s)
- Janusz Strzelczyk
- Department of Endocrinology and Neuroendocrine Tumors, Department of Pathophysiology and Endocrinology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Ksenia Janas
- Department of Endocrinology and Neuroendocrine Tumors, Department of Pathophysiology and Endocrinology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Joanna Katarzyna Strzelczyk
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Elżbieta Chełmecka
- Department of Medical Statistics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Poland
| | - Dariusz Kajdaniuk
- Department of Pathophysiology, Chair of Pathophysiology and Endocrinology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Beata Kos-Kudła
- Department of Endocrinology and Neuroendocrine Tumors, Department of Pathophysiology and Endocrinology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
9
|
Aydin H, Ozcelikkale A, Acar A. Exploiting Matrix Stiffness to Overcome Drug Resistance. ACS Biomater Sci Eng 2024; 10:4682-4700. [PMID: 38967485 PMCID: PMC11322920 DOI: 10.1021/acsbiomaterials.4c00445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
Drug resistance is arguably one of the biggest challenges facing cancer research today. Understanding the underlying mechanisms of drug resistance in tumor progression and metastasis are essential in developing better treatment modalities. Given the matrix stiffness affecting the mechanotransduction capabilities of cancer cells, characterization of the related signal transduction pathways can provide a better understanding for developing novel therapeutic strategies. In this review, we aimed to summarize the recent advancements in tumor matrix biology in parallel to therapeutic approaches targeting matrix stiffness and its consequences in cellular processes in tumor progression and metastasis. The cellular processes governed by signal transduction pathways and their aberrant activation may result in activating the epithelial-to-mesenchymal transition, cancer stemness, and autophagy, which can be attributed to drug resistance. Developing therapeutic strategies to target these cellular processes in cancer biology will offer novel therapeutic approaches to tailor better personalized treatment modalities for clinical studies.
Collapse
Affiliation(s)
- Hakan
Berk Aydin
- Department
of Biological Sciences, Middle East Technical
University, 06800, Ankara, Turkey
| | - Altug Ozcelikkale
- Department
of Mechanical Engineering, Middle East Technical
University, 06800, Ankara, Turkey
- Graduate
Program of Biomedical Engineering, Middle
East Technical University, 06800, Ankara, Turkey
| | - Ahmet Acar
- Department
of Biological Sciences, Middle East Technical
University, 06800, Ankara, Turkey
| |
Collapse
|
10
|
Zhong MZ, Xu MN, Zheng SQ, Cheng SQ, Zeng K, Huang XW. Manipulating host secreted protein gene expression: an indirect approach by HPV11/16 E6/E7 to suppress PBMC cytokine secretion. Virol J 2024; 21:172. [PMID: 39095779 PMCID: PMC11295672 DOI: 10.1186/s12985-024-02432-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/07/2024] [Indexed: 08/04/2024] Open
Abstract
Human papillomavirus (HPV) 11/16 E6/E7 proteins have been recognized to be pivotal in viral pathogenesis. This study sought to uncover the potential mechanisms of how HPV11/16 E6/E7-transfected keratinocytes inhibit cytokine secretion in peripheral blood mononuclear cells (PBMC). Upon co-culturing HPV11/16 E6/E7-transfected keratinocytes with PBMC in a non-contact manner, we observed a marked decrease in various cytokines secreted by PBMC. To determine if this suppression was mediated by specific common secreted factors, we conducted transcriptomic sequencing on these transfected cells. This analysis identified 53 common differentially secreted genes in all four HPV-transfected cells. Bioinformatics analysis demonstrated these genes were predominantly involved in immune regulation. Results from quantitative PCR (qPCR) and an extensive literature review suggested the downregulation of 12 genes (ACE2, BMP3, BPIFB1, CLU, CST6, CTF1, HMGB2, MMP12, PDGFA, RNASE7, SULF2, TGM2), and upregulation of 7 genes (CCL17, CCL22, FBLN1, PLAU, S100A7, S100A8, S100A9), may be crucial in modulating tumor immunity and combating pathogenic infections, with genes S100A8 and S100A9, and IL-17 signaling pathway being particularly noteworthy. Thus, HPV11/16 E6/E7 proteins may inhibit cytokine secretion of immune cells by altering the expression of host-secreted genes. Further exploration of these genes may yield new insights into the complex dynamics of HPV infection.
Collapse
Affiliation(s)
- Mei-Zhen Zhong
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mei-Nian Xu
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Si-Qi Zheng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shu-Qiong Cheng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kang Zeng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Xiao-Wen Huang
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
11
|
Lv S, Jiang H, Yu L, Zhang Y, Sun L, Xu J. SNX14 inhibits autophagy via the PI3K/AKT/mTOR signaling cascade in breast cancer cells. J Mol Histol 2024; 55:391-401. [PMID: 38869753 DOI: 10.1007/s10735-024-10209-1] [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: 09/06/2023] [Accepted: 06/01/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Sorting nexin 14 (SNX14) is a member of the sorting junction protein family. Its specific roles in cancer development remain unclear. Therefore, in this study, we aimed to determine the effects and underlying mechanisms of SNX14 on autophagy of breast cancer cells to aid in the therapeutic treatment of breast cancer. METHODS In this study, we performed in vitro experiments to determine the effect of SNX14 on breast cancer cell growth. Moreover, we used an MCF7 breast cancer tumor-bearing mouse model to confirm the effect of SNX14 on tumor cell growth in vivo. We also performed western blotting and quantitative polymerase chain reaction to identify the mechanism by which SNX14 affects breast cancer MCF7 cells. RESULTS We found that SNX14 regulated the onset and progression of breast cancer by promoting the proliferation and inhibiting the autophagy of MCF7 breast cancer cells. In vivo experiments further confirmed that SNX14 knockdown inhibited the tumorigenicity and inhibited the growth of tumor cells in tumor tissues of nude mice. In addition, western blotting analysis revealed that SNX14 modulate the autophagy of MCF7 breast cancer cells via the phosphoinositide 3-kinase/protein kinase B/mechanistic target of rapamycin kinase signaling pathway. CONCLUSION Our findings indicate that SNX14 is an essential tumor-promoting factor in the development of breast cancer.
Collapse
Affiliation(s)
- Sha Lv
- Department of Pharmacy, Zhejiang Hospital, Hangzhou, 310013, China
| | - Hongyan Jiang
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Lingyan Yu
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yafei Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Liangliang Sun
- Department of Pharmacy, Zhejiang Hospital, Hangzhou, 310013, China
| | - Junjun Xu
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
12
|
Yang N, Huang Y, Wang X, Wang D, Yao D, Ren G. Fibronectin-Targeting Dual-Modal MR/NIRF Imaging Contrast Agents for Diagnosis of Gastric Cancer and Peritoneal Metastasis. Bioconjug Chem 2024; 35:843-854. [PMID: 38775802 DOI: 10.1021/acs.bioconjchem.4c00208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
The prevalence and fatality rates of gastric cancer (GC) remain elevated, with advanced stages presenting a grim prognosis. Noninvasive diagnosis of GC cancer often proves challenging until the disease has progressed to an advanced stage or metastasized. Initially, the level of fibronectin (FN) in cancer-associated fibroblasts (CAFs) of GC was at least 3.7 times higher than that in normal fibroblasts. Herein, two FN-targeting magnetic resonance/near-infrared fluorescence (MR/NIRF) imaging contrast agents were developed to detect GC and peritoneal metastasis noninvasively. The probes CREKA-Cy7-(Gd-DOTA) and CREKA-Cy7-(Gd-DOTA)3 demonstrated significant FN-targeting capability (with dissociation constants of 1.0 and 2.1 mM) and effective MR imaging performance (with proton relaxivity values of 9.66 and 27.44 mM-1 s-1 at 9.4 T, 37 °C). In vivo imaging revealed a high signal-to-noise ratio and successful visualization of GC metastasis using NIRF imaging as well as successful tumor detection in MR imaging. Therefore, this study highlights the potential of FN-targeting probes for GC diagnosis and aids in the advancement of new diagnostic strategies for the clinical detection of GC.
Collapse
Affiliation(s)
- Ningxin Yang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yuelin Huang
- Shanghai University of Sport, Shanghai 200438, China
| | - Xiaoyu Wang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Dengbin Wang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Defan Yao
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Shanghai University of Sport, Shanghai 200438, China
| | - Gang Ren
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
13
|
Wei G, Chen X, Ruan T, Ma X, Zhu X, Wen W, He D, Tao K, Wu C. Human gastric cancer progression and stabilization of ATG2B through RNF5 binding facilitated by autophagy-associated CircDHX8. Cell Death Dis 2024; 15:410. [PMID: 38866787 PMCID: PMC11169566 DOI: 10.1038/s41419-024-06782-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/14/2024]
Abstract
The role of circDHX8 in the interplay between autophagy and gastric cancer (GC) progression remains unclear. In this study, we investigated the mechanism underlying the role of hsa_circ_003899 (circDHX8) in the malignancy of GC. Differential expression of circRNAs between GC and normal tissues was determined using circle-seq and microarray datasets (GSE83521). These circRNAs were validated using qPCR and Sanger sequencing. The function of circDHX8 was investigated through interference with circDHX8 expression experiments using in vitro and in vivo functional assays. Western blotting, immunofluorescence, and transmission electron microscopy were used to establish whether circDHX8 promoted autophagy in GC cells. To elucidate the mechanism underlying the circDHX8-mediated regulation of autophagy, we performed bioinformatics analysis, RNA pull-down, mass spectrometry (MS), RNA immunoprecipitation (RIP), and other western Blot related experiments. Hsa_circ_0003899 (circDHX8) was identified as upregulated and shown to enhance the malignant progression in GC cells by promoting cellular autophagy. Mechanistically, circDHX8 increased ATG2B protein levels by preventing ubiquitin-mediated degradation, thereby facilitating cell proliferation and invasion in GC. Additionally, circDHX8 directly interacts with the E3 ubiquitin-protein ligase RNF5, inhibiting the RNF5-mediated degradation of ATG2B. Concurrently, ATG2B, an acetylated protein, is subjected to SIRT1-mediated deacetylation, enhancing its binding to RNF5. Consequently, we established a novel mechanism for the role of circDHX8 in the malignant progression of GC.
Collapse
Affiliation(s)
- Guanxin Wei
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiang Chen
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tuo Ruan
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xianxiong Ma
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiuxian Zhu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wenhao Wen
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Danzeng He
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chuanqing Wu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
14
|
Ganesan K, Xu C, Xie C, Sui Y, Zheng C, Gao F, Chen J. Cryoprotective isoliquiritigenin-zein phosphatidylcholine nanoparticles inhibits breast cancer-bone metastasis by targeting JAK-STAT signaling pathways. Chem Biol Interact 2024; 396:111037. [PMID: 38719172 DOI: 10.1016/j.cbi.2024.111037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/27/2024] [Accepted: 05/05/2024] [Indexed: 05/19/2024]
Abstract
Breast cancer (BC) is the most common cancer in women and is known for its tendency to spread to the bones, causing significant health issues and mortality. In this study, we aimed to investigate whether cryoprotective isoliquiritigenin-zein phosphatidylcholine nanoparticles (ISL@ZLH NPs) could inhibit BC-induced bone destruction and tumor metastasis in both in vitro and animal models. To evaluate the potential of ISL@ZLH NPs, we conducted various experiments. First, we assessed cell viability, colony formation, transwell migration, and wound healing assays to determine the impact of ISL@ZLH NPs on BC cell behavior. Western blotting, TRAP staining and ALP activity were performed to examine the effects of ISL@ZLH NPs on osteoclast formation induced by MDA-MB-231 cell-conditioned medium and RANKL treated RAW 264.7 cells. Furthermore, we assessed the therapeutic impact of ISL@ZLH NPs on tumor-induced bone destruction using a mouse model of BC bone metastasis. Treatment with ISL@ZLH NPs effectively suppressed BC cell proliferation, colony formation, and motility, reducing their ability to metastasize. ISL@ZLH NPs significantly inhibited osteoclast formation and the expression of factors associated with bone destruction in BC cells. Additionally, ISL@ZLH NPs suppressed JAK-STAT signaling in RAW264.7 cells. In the BCBM mouse model, ISL@ZLH NPs led to a significant reduction in osteolytic bone lesions compared to the control group. Histological analysis and TRAP staining confirmed that ISL@ZLH NPs preserved the integrity of bone structure, preventing invasive metastasis by confining tumor growth to the bone marrow cavity. Furthermore, ISL@ZLH NPs effectively suppressed tumor-induced osteoclastogenesis, a key process in BC-related bone destruction. Our findings demonstrate that ISL@ZLH NPs have the potential to inhibit BC-induced bone destruction and tumor metastasis by targeting JAK-STAT signaling pathways and suppressing tumor-induced osteoclastogenesis. These results underscore the therapeutic promise of ISL@ZLH NPs in managing BC metastasis to the bones.
Collapse
Affiliation(s)
- Kumar Ganesan
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Cong Xu
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chunguang Xie
- Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yue Sui
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chuan Zheng
- Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Fei Gao
- Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Jianping Chen
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
15
|
Ferraresi A, Girone C, Maheshwari C, Vallino L, Dhanasekaran DN, Isidoro C. Ovarian Cancer Cell-Conditioning Medium Induces Cancer-Associated Fibroblast Phenoconversion through Glucose-Dependent Inhibition of Autophagy. Int J Mol Sci 2024; 25:5691. [PMID: 38891879 PMCID: PMC11171902 DOI: 10.3390/ijms25115691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/19/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
One aspect of ovarian tumorigenesis which is still poorly understood is the tumor-stroma interaction, which plays a major role in chemoresistance and tumor progression. Cancer-associated fibroblasts (CAFs), the most abundant stromal cell type in the tumor microenvironment, influence tumor growth, metabolism, metastasis, and response to therapy, making them attractive targets for anti-cancer treatment. Unraveling the mechanisms involved in CAFs activation and maintenance is therefore crucial for the improvement of therapy efficacy. Here, we report that CAFs phenoconversion relies on the glucose-dependent inhibition of autophagy. We show that ovarian cancer cell-conditioning medium induces a metabolic reprogramming towards the CAF-phenotype that requires the autophagy-dependent glycolytic shift. In fact, 2-deoxy-D-glucose (2DG) strongly hampers such phenoconversion and, most importantly, induces the phenoreversion of CAFs into quiescent fibroblasts. Moreover, pharmacological inhibition (by proline) or autophagy gene knockdown (by siBECN1 or siATG7) promotes, while autophagy induction (by either 2DG or rapamycin) counteracts, the metabolic rewiring induced by the ovarian cancer cell secretome. Notably, the nutraceutical resveratrol (RV), known to inhibit glucose metabolism and to induce autophagy, promotes the phenoreversion of CAFs into normal fibroblasts even in the presence of ovarian cancer cell-conditioning medium. Overall, our data support the view of testing autophagy inducers for targeting the tumor-promoting stroma as an adjuvant strategy to improve therapy success rates, especially for tumors with a highly desmoplastic stroma, like ovarian cancer.
Collapse
Affiliation(s)
- Alessandra Ferraresi
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.G.); (C.M.); (L.V.)
| | - Carlo Girone
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.G.); (C.M.); (L.V.)
| | - Chinmay Maheshwari
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.G.); (C.M.); (L.V.)
| | - Letizia Vallino
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.G.); (C.M.); (L.V.)
| | - Danny N. Dhanasekaran
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.G.); (C.M.); (L.V.)
| |
Collapse
|
16
|
Went M, Sud A, Mills C, Hyde A, Culliford R, Law P, Vijayakrishnan J, Gockel I, Maj C, Schumacher J, Palles C, Kaiser M, Houlston R. Phenome-wide Mendelian randomisation analysis of 378,142 cases reveals risk factors for eight common cancers. Nat Commun 2024; 15:2637. [PMID: 38527997 PMCID: PMC10963765 DOI: 10.1038/s41467-024-46927-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 03/08/2024] [Indexed: 03/27/2024] Open
Abstract
For many cancers there are only a few well-established risk factors. Here, we use summary data from genome-wide association studies (GWAS) in a Mendelian randomisation (MR) phenome-wide association study (PheWAS) to identify potentially causal relationships for over 3,000 traits. Our outcome datasets comprise 378,142 cases across breast, prostate, colorectal, lung, endometrial, oesophageal, renal, and ovarian cancers, as well as 485,715 controls. We complement this analysis by systematically mining the literature space for supporting evidence. In addition to providing supporting evidence for well-established risk factors (smoking, alcohol, obesity, lack of physical activity), we also find sex steroid hormones, plasma lipids, and telomere length as determinants of cancer risk. A number of the molecular factors we identify may prove to be potential biomarkers. Our analysis, which highlights aetiological similarities and differences in common cancers, should aid public health prevention strategies to reduce cancer burden. We provide a R/Shiny app to visualise findings.
Collapse
Affiliation(s)
- Molly Went
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK.
| | - Amit Sud
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Charlie Mills
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Abi Hyde
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- Department of Engineering, University of Cambridge, Cambridge, UK
| | - Richard Culliford
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Philip Law
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | | | - Ines Gockel
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Carlo Maj
- Center for Human Genetics, University Hospital of Marburg, Marburg, Germany
| | | | - Claire Palles
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Martin Kaiser
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Richard Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| |
Collapse
|
17
|
Yu T, Rui L, Jiumei Z, Ziwei L, Ying H. Advances in the study of autophagy in breast cancer. Breast Cancer 2024; 31:195-204. [PMID: 38315272 PMCID: PMC10901946 DOI: 10.1007/s12282-023-01541-7] [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: 11/13/2023] [Accepted: 12/25/2023] [Indexed: 02/07/2024]
Abstract
Breast cancer is the most prevalent malignant tumor among women, with a high incidence and mortality rate all year round, which seriously affects women's health. Autophagy, a well-conserved cellular process inherent in eukaryotic organisms, plays a pivotal role in degrading damaged proteins and organelles, recycling their breakdown products to aid cells in navigating stress and gradually restoring homeostatic equilibrium. Recent studies have unveiled the intricate connection between autophagy and breast cancer. Autophagy is a double-edged sword in breast cancer, demonstrating a dual role: restraining its onset and progression on one hand, while promoting its metastasis and advancement on the other. It is also because of this interrelationship between the two that regulation of autophagy in the treatment of breast cancer is now an important strategy in clinical treatment. In this article, we systematically survey the recent research findings, elucidating the multifaceted role of autophagy in breast cancer and its underlying mechanisms, with the aim of contributing new references to the clinical management of breast cancer.
Collapse
Affiliation(s)
- Tang Yu
- The Third Affiliated Hospital of Kunming Medical University, Kunming, China
- The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liu Rui
- The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhao Jiumei
- Chongqing Nanchuan District People's Hospital, Chongqing, China
| | - Li Ziwei
- Chongqing Health Center for Women and Children, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Hu Ying
- The Second Affiliatied Hospital of Kunming Medical University and Department of Clinical Larboratory, Kunming, China.
| |
Collapse
|
18
|
Pulat S, Yang I, Lee J, Hwang S, Zhou R, Gamage CDB, Varlı M, Taş İ, Yang Y, Park SY, Hong A, Kim JH, Oh DC, Kim H, Nam SJ, Kang H. Anithiactin D, a Phenylthiazole Natural Product from Mudflat-Derived Streptomyces sp., Suppresses Motility of Cancer Cells. Mar Drugs 2024; 22:88. [PMID: 38393059 PMCID: PMC10889970 DOI: 10.3390/md22020088] [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: 01/15/2024] [Revised: 02/02/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Anithiactin D (1), a 2-phenylthiazole class of natural products, was isolated from marine mudflat-derived actinomycetes Streptomyces sp. 10A085. The chemical structure of 1 was elucidated based on the interpretation of NMR and MS data. The absolute configuration of 1 was determined by comparing the experimental and calculated electronic circular dichroism (ECD) spectral data. Anithiactin D (1) significantly decreased cancer cell migration and invasion activities at a concentration of 5 μM via downregulation of the epithelial-to-mesenchymal transition (EMT) markers in A549, AGS, and Caco-2 cell lines. Moreover, 1 inhibited the activity of Rho GTPases, including Rac1 and RhoA in the A549 cell line, suppressed RhoA in AGS and Caco-2 cell lines, and decreased the mRNA expression levels of some matrix metalloproteinases (MMPs) in AGS and Caco-2 cell lines. Thus 1, which is a new entity of the 2-phenylthiazole class of natural products with a unique aniline-indole fused moiety, is a potent inhibitor of the motility of cancer cells.
Collapse
Affiliation(s)
- Sultan Pulat
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea; (S.P.); (R.Z.); (C.D.B.G.); (M.V.); (İ.T.); (Y.Y.); (S.-Y.P.)
| | - Inho Yang
- Department of Convergence Study on the Ocean Science and Technology, Korea Maritime and Ocean University, Busan 49112, Republic of Korea;
| | - Jihye Lee
- Laboratories of Marine New Drugs, REDONE Seoul, Seoul 08594, Republic of Korea;
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Republic of Korea; (A.H.); (J.-H.K.)
| | - Sunghoon Hwang
- Natural Products Research Institute, College of Pharmacy, Seoul National University, NS-80, Seoul 08826, Republic of Korea; (S.H.); (D.-C.O.)
| | - Rui Zhou
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea; (S.P.); (R.Z.); (C.D.B.G.); (M.V.); (İ.T.); (Y.Y.); (S.-Y.P.)
| | - Chathurika D. B. Gamage
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea; (S.P.); (R.Z.); (C.D.B.G.); (M.V.); (İ.T.); (Y.Y.); (S.-Y.P.)
| | - Mücahit Varlı
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea; (S.P.); (R.Z.); (C.D.B.G.); (M.V.); (İ.T.); (Y.Y.); (S.-Y.P.)
| | - İsa Taş
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea; (S.P.); (R.Z.); (C.D.B.G.); (M.V.); (İ.T.); (Y.Y.); (S.-Y.P.)
| | - Yi Yang
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea; (S.P.); (R.Z.); (C.D.B.G.); (M.V.); (İ.T.); (Y.Y.); (S.-Y.P.)
| | - So-Yeon Park
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea; (S.P.); (R.Z.); (C.D.B.G.); (M.V.); (İ.T.); (Y.Y.); (S.-Y.P.)
| | - Ahreum Hong
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Republic of Korea; (A.H.); (J.-H.K.)
| | - Jeong-Hyeon Kim
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Republic of Korea; (A.H.); (J.-H.K.)
| | - Dong-Chan Oh
- Natural Products Research Institute, College of Pharmacy, Seoul National University, NS-80, Seoul 08826, Republic of Korea; (S.H.); (D.-C.O.)
| | - Hangun Kim
- College of Pharmacy, Sunchon National University, Sunchon 57922, Republic of Korea; (S.P.); (R.Z.); (C.D.B.G.); (M.V.); (İ.T.); (Y.Y.); (S.-Y.P.)
| | - Sang-Jip Nam
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Republic of Korea; (A.H.); (J.-H.K.)
| | - Heonjoong Kang
- Laboratory of Marine Drugs, School of Earth and Environmental Sciences, Seoul National University, NS-80, Seoul 08826, Republic of Korea
| |
Collapse
|
19
|
Yan J, Xiao G, Yang C, Liu Q, Lv C, Yu X, Zhou Z, Lin S, Bai Z, Lin H, Zhang R, Liu C. Cancer-Associated Fibroblasts Promote Lymphatic Metastasis in Cholangiocarcinoma via the PDGF-BB/PDGFR-β Mediated Paracrine Signaling Network. Aging Dis 2024; 15:369-389. [PMID: 37307823 PMCID: PMC10796099 DOI: 10.14336/ad.2023.0420] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 04/20/2023] [Indexed: 06/14/2023] Open
Abstract
Patients with cholangiocarcinoma (CCA) with lymph node metastasis (LNM) have the worst prognosis, even after complete resection; however, the underlying mechanism remains unclear. Here, we established CAF-derived PDGF-BB as a regulator of LMN in CCA. Proteomics analysis revealed upregulation of PDGF-BB in CAFs derived from patients with CCA with LMN (LN+CAFs). Clinically, the expression of CAF-PDGF-BB correlated with poor prognosis and increased LMN in patients with CCA, while CAF-secreted PDGF-BB enhanced lymphatic endothelial cell (LEC)-mediated lymphangiogenesis and promoted the trans-LEC migration ability of tumor cells. Co-injection of LN+CAFs and cancer cells increased tumor growth and LMN in vivo. Mechanistically, CAF-derived PDGF-BB activated its receptor PDGFR-β and its downstream ERK1/2-JNK signaling pathways in LECs to promote lymphoangiogenesis, while it also upregulated the PDGFR-β-GSK-P65-mediated tumor cell migration. Finally, targeting PDGF-BB/PDGFR-β or the GSK-P65 signaling axis prohibited CAF-mediated popliteal lymphatic metastasis (PLM) in vivo. Overall, our findings revealed that CAFs promote tumor growth and LMN via a paracrine network, identifying a promising therapeutic target for patients with advanced CCA.
Collapse
Affiliation(s)
- Jian Yan
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Gang Xiao
- Department of Thoracic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China.
- Center for Medical Research on Innovation and Translation, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China.
| | - Caini Yang
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Qinqin Liu
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Cui Lv
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xianhuan Yu
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Ziyu Zhou
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Shusheng Lin
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Zhenhua Bai
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Haoming Lin
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Rui Zhang
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Chao Liu
- Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
20
|
Sari D, Gozuacik D, Akkoc Y. Role of autophagy in cancer-associated fibroblast activation, signaling and metabolic reprograming. Front Cell Dev Biol 2024; 11:1274682. [PMID: 38234683 PMCID: PMC10791779 DOI: 10.3389/fcell.2023.1274682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/08/2023] [Indexed: 01/19/2024] Open
Abstract
Tumors not only consist of cancerous cells, but they also harbor several normal-like cell types and non-cellular components. cancer-associated fibroblasts (CAFs) are one of these cellular components that are found predominantly in the tumor stroma. Autophagy is an intracellular degradation and quality control mechanism, and recent studies provided evidence that autophagy played a critical role in CAF formation, metabolic reprograming and tumor-stroma crosstalk. Therefore, shedding light on the autophagy and its role in CAF biology might help us better understand the roles of CAFs and the TME in cancer progression and may facilitate the exploitation of more efficient cancer diagnosis and treatment. Here, we provide an overview about the involvement of autophagy in CAF-related pathways, including transdifferentiation and activation of CAFs, and further discuss the implications of targeting tumor stroma as a treatment option.
Collapse
Affiliation(s)
- Dyana Sari
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| | - Devrim Gozuacik
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
- Department of Medical Biology, School of Medicine, Koç University, Istanbul, Türkiye
- Department of Biotechnology, SUNUM Nanotechnology Research and Application Center, Istanbul, Türkiye
| | - Yunus Akkoc
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| |
Collapse
|
21
|
Hu Y, Wang H, Liu Y. NETosis: Sculpting tumor metastasis and immunotherapy. Immunol Rev 2024; 321:263-279. [PMID: 37712361 DOI: 10.1111/imr.13277] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/16/2023]
Abstract
The process of neutrophil extracellular traps (NETs) formation, called NETosis, is a peculiar death modality of neutrophils, which was first observed as an immune response against bacterial infection. However, recent work has revealed the unique biology of NETosis in facilitating tumor metastatic process. Neutrophil extracellular traps released by the tumor microenvironment (TME) shield tumor cells from cytotoxic immunity, leading to impaired tumor clearance. Besides, tumor cells tapped by NETs enable to travel through vessels and subsequently seed distant organs. Targeted ablation of NETosis has been proven to be beneficial in potentiating the efficacy of cancer immunotherapy in the metastatic settings. This review outlines the impact of NETosis at almost all stages of tumor metastasis. Furthermore, understanding the multifaceted interplay between NETosis and the TME components is crucial for supporting the rational development of highly effective combination immunotherapeutic strategies with anti-NETosis for patients with metastatic disease.
Collapse
Affiliation(s)
- Yanyan Hu
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Houhong Wang
- Department of General Surgery, The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, China
| | - Yang Liu
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, China
| |
Collapse
|
22
|
Zhang W, Wang J, Liu C, Li Y, Sun C, Wu J, Wu Q. Crosstalk and plasticity driving between cancer-associated fibroblasts and tumor microenvironment: significance of breast cancer metastasis. J Transl Med 2023; 21:827. [PMID: 37978384 PMCID: PMC10657029 DOI: 10.1186/s12967-023-04714-2] [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: 08/18/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) are the most abundant stromal cell population in breast tumors. A functionally diverse population of CAFs increases the dynamic complexity of the tumor microenvironment (TME). The intertwined network of the TME facilitates the interaction between activated CAFs and breast cancer cells, which can lead to the proliferation and invasion of breast cells. Considering the special transmission function of CAFs, the aim of this review is to summarize and highlight the crosstalk between CAFs and breast cancer cells in the TME as well as the relationship between CAFs and extracellular matrix (ECM), soluble cytokines, and other stromal cells in the metastatic state. The crosstalk between cancer-associated fibroblasts and tumor microenvironment also provides a plastic therapeutic target for breast cancer metastasis. In the course of the study, the inhibitory effects of different natural compounds on targeting CAFs and the advantages of different drug combinations were summarized. CAFs are also widely used in the diagnosis and treatment of breast cancer. The cumulative research on this phenomenon supports the establishment of a targeted immune microenvironment as a possible breakthrough in the prevention of invasive metastasis of breast cancer.
Collapse
Affiliation(s)
- Wenfeng Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau, China
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, 261000, China
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Jia Wang
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau, China
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Cun Liu
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, 261000, China
| | - Ye Li
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau, China
| | - Changgang Sun
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau, China.
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, 261000, China.
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, China.
| | - Jibiao Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Qibiao Wu
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau, China.
| |
Collapse
|
23
|
Went M, Sud A, Mills C, Hyde A, Culliford R, Law P, Vijayakrishnan J, Gockel I, Maj C, Schumacher J, Palles C, Kaiser M, Houlston R. Risk factors for eight common cancers revealed from a phenome-wide Mendelian randomisation analysis of 378,142 cases and 485,715 controls. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.15.23285952. [PMID: 37066289 PMCID: PMC10104236 DOI: 10.1101/2023.02.15.23285952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
For many cancers there are few well-established risk factors. Summary data from genome-wide association studies (GWAS) can be used in a Mendelian randomisation (MR) phenome-wide association study (PheWAS) to identify causal relationships. We performed a MR-PheWAS of breast, prostate, colorectal, lung, endometrial, oesophageal, renal, and ovarian cancers, comprising 378,142 cases and 485,715 controls. To derive a more comprehensive insight into disease aetiology we systematically mined the literature space for supporting evidence. We evaluated causal relationships for over 3,000 potential risk factors. In addition to identifying well-established risk factors (smoking, alcohol, obesity, lack of physical activity), we provide evidence for specific factors, including dietary intake, sex steroid hormones, plasma lipids and telomere length as determinants of cancer risk. We also implicate molecular factors including plasma levels of IL-18, LAG-3, IGF-1, CT-1, and PRDX1 as risk factors. Our analyses highlight the importance of risk factors that are common to many cancer types but also reveal aetiological differences. A number of the molecular factors we identify have the potential to be biomarkers. Our findings should aid public health prevention strategies to reduce cancer burden. We provide a R/Shiny app (https://mrcancer.shinyapps.io/mrcan/) to visualise findings.
Collapse
Affiliation(s)
- Molly Went
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Amit Sud
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- Haemato-oncology Unit, The Royal Marsden Hospital NHS Foundation Trust, Sutton, UK
| | - Charlie Mills
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Abi Hyde
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Richard Culliford
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Philip Law
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | | | - Ines Gockel
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Carlo Maj
- Center for Human Genetics, University Hospital of Marburg, Marburg, Germany
| | | | - Claire Palles
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Martin Kaiser
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- Haemato-oncology Unit, The Royal Marsden Hospital NHS Foundation Trust, Sutton, UK
| | - Richard Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| |
Collapse
|
24
|
Went M, Sud A, Mills C, Hyde A, Culliford R, Law P, Vijayakrishnan J, Gockel I, Maj C, Schumacher J, Palles C, Kaiser M, Houlston R. Risk factors for eight common cancers revealed from a phenome-wide Mendelian randomisation analysis of 378,142 cases and 485,715 controls. RESEARCH SQUARE 2023:rs.3.rs-2587058. [PMID: 36993383 PMCID: PMC10055507 DOI: 10.21203/rs.3.rs-2587058/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
For many cancers there are few well-established risk factors. Summary data from genome-wide association studies (GWAS) can be used in a Mendelian randomisation (MR) phenome-wide association study (PheWAS) to identify causal relationships. We performed a MR-PheWAS of breast, prostate, colorectal, lung, endometrial, oesophageal, renal, and ovarian cancers, comprising 378,142 cases and 485,715 controls. To derive a more comprehensive insight into disease aetiology we systematically mined the literature space for supporting evidence. We evaluated causal relationships for over 3,000 potential risk factors. In addition to identifying well-established risk factors (smoking, alcohol, obesity, lack of physical activity), we provide evidence for specific factors, including dietary intake, sex steroid hormones, plasma lipids and telomere length as determinants of cancer risk. We also implicate molecular factors including plasma levels of IL-18, LAG-3, IGF-1, CT-1, and PRDX1 as risk factors. Our analyses highlight the importance of risk factors that are common to many cancer types but also reveal aetiological differences. A number of the molecular factors we identify have the potential to be biomarkers. Our findings should aid public health prevention strategies to reduce cancer burden. We provide a R/Shiny app (https://mrcancer.shinyapps.io/mrcan/) to visualise findings.
Collapse
Affiliation(s)
- Molly Went
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Amit Sud
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- Haemato-oncology Unit, The Royal Marsden Hospital NHS Foundation Trust, Sutton, UK
| | - Charlie Mills
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Abi Hyde
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Richard Culliford
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Philip Law
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | | | - Ines Gockel
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| | - Carlo Maj
- Center for Human Genetics, University Hospital of Marburg, Marburg, Germany
| | | | - Claire Palles
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Martin Kaiser
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- Haemato-oncology Unit, The Royal Marsden Hospital NHS Foundation Trust, Sutton, UK
| | - Richard Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| |
Collapse
|
25
|
The Role of Autophagy in Breast Cancer Metastasis. Biomedicines 2023; 11:biomedicines11020618. [PMID: 36831154 PMCID: PMC9953203 DOI: 10.3390/biomedicines11020618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/07/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Patient morbidity and mortality is significantly increased in metastatic breast cancer. The metastasis process of breast cancer is very complicated and is delicately controlled by various factors. Autophagy is one of the important regulatory factors affecting metastasis in breast cancer by engaging in cell mobility, metabolic adaptation, tumor dormancy, and cancer stem cells. Here, we discuss the effects of autophagy on metastasis in breast cancer and assess the potential use of autophagy modulators for metastasis treatment.
Collapse
|
26
|
Novel Anti-Cancer Products Targeting AMPK: Natural Herbal Medicine against Breast Cancer. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020740. [PMID: 36677797 PMCID: PMC9863744 DOI: 10.3390/molecules28020740] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/01/2023] [Accepted: 01/04/2023] [Indexed: 01/15/2023]
Abstract
Breast cancer is a common cancer in women worldwide. The existing clinical treatment strategies have been able to limit the progression of breast cancer and cancer metastasis, but abnormal metabolism, immunosuppression, and multidrug resistance involving multiple regulators remain the major challenges for the treatment of breast cancer. Adenosine 5'-monophosphate (AMP)-Activated Protein Kinase (AMPK) can regulate metabolic reprogramming and reverse the "Warburg effect" via multiple metabolic signaling pathways in breast cancer. Previous studies suggest that the activation of AMPK suppresses the growth and metastasis of breast cancer cells, as well as stimulating the responses of immune cells. However, some other reports claim that the development and poor prognosis of breast cancer are related to the overexpression and aberrant activation of AMPK. Thus, the role of AMPK in the progression of breast cancer is still controversial. In this review, we summarize the current understanding of AMPK, particularly the comprehensive bidirectional functions of AMPK in cancer progression; discuss the pharmacological activators of AMPK and some specific molecules, including the natural products (including berberine, curcumin, (-)-epigallocatechin-3-gallate, ginsenosides, and paclitaxel) that influence the efficacy of these activators in cancer therapy; and elaborate the role of AMPK as a potential therapeutic target for the treatment of breast cancer.
Collapse
|