1
|
Kahkesh S, Hedayati N, Rahimzadeh P, Farahani N, Khoozani MF, Abedi M, Nabavi N, Naeimi B, Khoshnazar SM, Alimohammadi M, Alaei E, Mahmoodieh B. The function of circular RNAs in regulating Wnt/β-catenin signaling: An innovative therapeutic strategy for breast and gynecological cancers. Pathol Res Pract 2025; 270:155944. [PMID: 40228402 DOI: 10.1016/j.prp.2025.155944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/16/2025]
Abstract
Breast cancer (BC) and gynecological malignancies, including cervical, ovarian, and uterine cancers, are significant global health challenges due to their high prevalence, complex nature, and elevated mortality rates. Dysregulation of the Wnt/β-catenin signaling pathway is a common feature in gynecological malignancies, contributing to cancer cell growth, progression, migration, and metastasis. Recent studies have highlighted the pivotal role of non-coding RNAs (ncRNAs), particularly circular RNAs (circRNAs), in modulating the Wnt/β-catenin signaling pathway. Acting as sponges for microRNAs (miRNAs), circRNAs regulate key oncogenic and tumor-suppressive processes by influencing Wnt-related components. This research explores the role of circRNAs in breast and gynecological malignancies, focusing on their regulatory effects on the Wnt/β-catenin pathway. The findings reveal that circRNAs modulate critical cellular processes such as proliferation, apoptosis, autophagy, and metastasis, with potential implications for therapeutic interventions. Targeting circRNA-mediated dysregulation of Wnt signaling could offer novel strategies for improving diagnostic precision, treatment efficacy, and survival outcomes in breast and gynecological cancers.
Collapse
Affiliation(s)
- Samaneh Kahkesh
- Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahdi Farhadi Khoozani
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Maryam Abedi
- Department of Pathology, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, Canada
| | - Bita Naeimi
- Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
| | - Seyedeh Mahdieh Khoshnazar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Elmira Alaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Behnaz Mahmoodieh
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
2
|
Bischof K, Cremaschi A, Eroukhmanoff L, Landskron J, Flage‐Larsen L, Gade A, Bjørge L, Urbanucci A, Taskén K. Patient-derived acellular ascites fluid affects drug responses in ovarian cancer cell lines through the activation of key signalling pathways. Mol Oncol 2025; 19:81-98. [PMID: 39245677 PMCID: PMC11705723 DOI: 10.1002/1878-0261.13726] [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: 03/04/2024] [Revised: 07/03/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024] Open
Abstract
Malignant ascites is commonly produced in advanced epithelial ovarian cancer (EOC) and serves as unique microenvironment for tumour cells. Acellular ascites fluid (AAF) is rich in signalling molecules and has been proposed to play a role in the induction of chemoresistance. Through in vitro testing of drug sensitivity and by assessing intracellular phosphorylation status in response to mono- and combination treatment of five EOC cell lines after incubation with AAFs derived from 20 different patients, we investigated the chemoresistance-inducing potential of ascites. We show that the addition of AAFs to the culture media of EOC cell lines has the potential to induce resistance to standard-of-care drugs (SCDs). We also show that AAFs induce time- and concentration-dependent activation of downstream signalling to signal transducer and activator of transcription 3 (STAT3), and concomitantly altered phosphorylation of mitogen-activated protein kinase kinase (MEK), phosphoinositide 3-kinase (PI3K)-protein kinase B (AKT) and nuclear factor NF-kappa-B (NFκB). Antibodies targeting the interleukin-6 receptor (IL6R) effectively blocked phosphorylation of STAT3 and STAT1. Treatments with SCDs were effective in reducing cell viability in only a third of 30 clinically relevant conditions examined, defined as combinations of drugs, different cell lines and AAFs. Combinations of SCDs and novel therapeutics such as trametinib, fludarabine or rapamycin were superior in another third. Notably, we could nominate effective treatment combinations in almost all conditions except in 4 out of 30 conditions, in which trametinib or fludarabine showed higher efficacy alone. Taken together, our study underscores the importance of the molecular characterisation of individual patients' AAFs and the impact on treatment resistance as providing clinically meaningful information for future precision treatment approaches in EOC.
Collapse
Affiliation(s)
- Katharina Bischof
- Department of Cancer Immunology, Institute for Cancer ResearchUniversity of OsloNorway
- Division of Cancer Medicine, Department of Gynecological OncologyOslo University HospitalNorway
| | - Andrea Cremaschi
- Centre for Molecular Medicine Norway (NCMM)Nordic EMBL Partnership, University of OsloNorway
- Oslo Centre for Biostatistics and EpidemiologyUniversity of OsloNorway
- Singapore Institute for Clinical Sciences, A*STARSingapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore
| | - Lena Eroukhmanoff
- Centre for Molecular Medicine Norway (NCMM)Nordic EMBL Partnership, University of OsloNorway
| | - Johannes Landskron
- Centre for Molecular Medicine Norway (NCMM)Nordic EMBL Partnership, University of OsloNorway
| | - Lise‐Lotte Flage‐Larsen
- Centre for Molecular Medicine Norway (NCMM)Nordic EMBL Partnership, University of OsloNorway
| | - Alexandra Gade
- Centre for Molecular Medicine Norway (NCMM)Nordic EMBL Partnership, University of OsloNorway
| | - Line Bjørge
- Department of Obstetrics and GynaecologyHaukeland University HospitalBergenNorway
- Department of Clinical Science, Centre for Cancer Biomarkers CCBIOUniversity of BergenNorway
| | - Alfonso Urbanucci
- Faculty of Medicine and Health TechnologyTAYS Cancer Centre and FICAN Mid, Tampere UniversityFinland
- Department of Tumor Biology, Institute for Cancer ResearchUniversity of OsloNorway
| | - Kjetil Taskén
- Department of Cancer Immunology, Institute for Cancer ResearchUniversity of OsloNorway
- Institute of Clinical MedicineUniversity of OsloNorway
| |
Collapse
|
3
|
Zhu Y, He Y, Gan R. Wnt Signaling in Hepatocellular Carcinoma: Biological Mechanisms and Therapeutic Opportunities. Cells 2024; 13:1990. [PMID: 39682738 PMCID: PMC11640042 DOI: 10.3390/cells13231990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/19/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
Hepatocellular carcinoma (HCC), characterized by significant morbidity and mortality rates, poses a substantial threat to human health. The expression of ligands and receptors within the classical and non-classical Wnt signaling pathways plays an important role in HCC. The Wnt signaling pathway is essential for regulating multiple biological processes in HCC, including proliferation, invasion, migration, tumor microenvironment modulation, epithelial-mesenchymal transition (EMT), stem cell characteristics, and autophagy. Molecular agents that specifically target the Wnt signaling pathway have demonstrated significant potential for the treatment of HCC. However, the precise mechanism by which the Wnt signaling pathway interacts with HCC remains unclear. In this paper, we review the alteration of the Wnt signaling pathway in HCC, the mechanism of Wnt pathway action in HCC, and molecular agents targeting the Wnt pathway. This paper provides a theoretical foundation for identifying molecular agents targeting the Wnt pathway in hepatocellular carcinoma.
Collapse
Affiliation(s)
| | | | - Runliang Gan
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang 421001, China; (Y.Z.); (Y.H.)
| |
Collapse
|
4
|
Huang Y, Fan M, Liu Y, Jiang X, Du K, Wu A, Li Q, Wu Y, Liang J, Wang K. Novel biomarkers and drug correlations of non-canonical WNT signaling in prostate and breast cancer. Discov Oncol 2024; 15:511. [PMID: 39347881 PMCID: PMC11442966 DOI: 10.1007/s12672-024-01394-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024] Open
Abstract
Prostate cancer (PCa) and breast cancer (BC) present formidable challenges in global cancer-related mortality, necessitating effective management strategies. The present study explores non-canonical Wnt signaling in PCa and BC, aiming to identify biomarkers and assess their clinical and therapeutic implications. Co-expression analyses reveal distinct gene patterns, with five overlapping genes (SULF1, ALG3, IL16, PLXNA2 and RASGFR2) exhibiting divergent expression in both cancers. Clinical relevance investigations demonstrate correlations with TNM stages and biochemical recurrence. Drug correlation analyses unveil potential therapeutic avenues, indicating that Wnt5a and ROR2 expressions are related to MEK inhibitor sensitivity in cancers. Meanwhile, further correlation analyses were conducted between drugs and the other novel non-canonical WNT genes (ALG3, IL16, SULF1, PLXNA2, and RASGRF2). Our findings contribute to understanding non-canonical Wnt signaling, offering insights into cancer progression and potential personalized treatment approaches.
Collapse
Affiliation(s)
- Yongming Huang
- Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Meiyin Fan
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yushuai Liu
- Ophthalmology Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaoying Jiang
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | | | | | - Qingyi Li
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yingying Wu
- Department of Mathematics, University of Houston, Houston, USA.
| | - Jiaqian Liang
- Department of Urology, Wuhan No.1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Keshan Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
5
|
Huang J, Tan R. HMOX1: A pivotal regulator of prognosis and immune dynamics in ovarian cancer. BMC Womens Health 2024; 24:476. [PMID: 39210460 PMCID: PMC11363456 DOI: 10.1186/s12905-024-03309-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND This study investigates the intricate role of Heme Oxygenase 1 (HMOX1) in ovarian cancer, emphasizing its prognostic significance, influence on immune cell infiltration, and impact on the malignant characteristics of primary ovarian cancer cells. MATERIALS AND METHODS Our research began with an analysis of HMOX1 expression and its prognostic implications using data from The Cancer Genome Atlas (TCGA) dataset, supported by immunohistochemical staining. Further analyses encompassed co-expression studies, Gene Ontology (GO) annotations, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment. We utilized the TIMER and TISIDB platforms to evaluate the immunotherapeutic potential of HMOX1. Additionally, in vitro studies that involved modulating HMOX1 levels in primary ovarian cancer cells were conducted to confirm its biological functions. RESULTS Our findings indicate a significant overexpression of HMOX1 in ovarian cancer, which correlates with increased tumor malignancy and poorer prognosis. HMOX1 was shown to significantly modulate the infiltration of immune cells, particularly neutrophils and macrophages. Single-cell RNA sequencing (scRNA-seq) analysis revealed that HMOX1 is predominantly expressed in tumor-associated macrophages (TAMs), with a positive correlation to chemokines and their receptors. An increase in HMOX1 levels was associated with heightened levels of immunoinhibitors, immunostimulators, and MHC molecules. Functional assays demonstrated that HMOX1 knockdown promotes apoptosis, attenuating cell proliferation and invasion, while its overexpression yields opposing effects. CONCLUSION HMOX1 emerges as a critical therapeutic target, intricately involved in immunomodulation, prognosis, and the malignant behavior of ovarian cancer. This highlights HMOX1 as a potential biomarker and therapeutic target in the fight against ovarian cancer.
Collapse
Affiliation(s)
- Jinfa Huang
- Department of Gynecology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong, 528308, China
| | - Ruiwan Tan
- Department of Ultrasound, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong, 528308, China.
| |
Collapse
|
6
|
Chehade H, Gogoi R, Adzibolosu NK, Galoforo S, Fehmi RA, Kheil M, Fox A, Kim S, Rattan R, Hou Z, Morris RT, Matherly LH, Mor G, Alvero AB. BRCA Status Dictates Wnt Responsiveness in Epithelial Ovarian Cancer. CANCER RESEARCH COMMUNICATIONS 2024; 4:2075-2088. [PMID: 39028933 PMCID: PMC11320024 DOI: 10.1158/2767-9764.crc-24-0111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/17/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024]
Abstract
The association of BRCA1 and BRCA2 mutations with increased risk for developing epithelial ovarian cancer is well established. However, the observed clinical differences, particularly the improved therapy response and patient survival in BRCA2-mutant patients, are unexplained. Our objective is to identify molecular pathways that are differentially regulated upon the loss of BRCA1 and BRCA2 functions in ovarian cancer. Transcriptomic and pathway analyses comparing BRCA1-mutant, BRCA2-mutant, and homologous recombination wild-type ovarian tumors showed differential regulation of the Wnt/β-catenin pathway. Using Wnt3A-treated BRCA1/2 wild-type, BRCA1-null, and BRCA2-null mouse ovarian cancer cells, we observed preferential activation of canonical Wnt/β-catenin signaling in BRCA1/2 wild-type ovarian cancer cells, whereas noncanonical Wnt/β-catenin signaling was preferentially activated in the BRCA1-null ovarian cancer cells. Interestingly, BRCA2-null mouse ovarian cancer cells demonstrated a unique response to Wnt3A with the preferential upregulation of the Wnt signaling inhibitor Axin2. In addition, decreased phosphorylation and enhanced stability of β-catenin were observed in BRCA2-null mouse ovarian cancer cells, which correlated with increased inhibitory phosphorylation on GSK3β. These findings open venues for the translation of these molecular observations into modalities that can impact patient survival. SIGNIFICANCE We show that BRCA1 and BRCA2 mutation statuses differentially impact the regulation of the Wnt/β-catenin signaling pathway, a major effector of cancer initiation and progression. Our findings provide a better understanding of molecular mechanisms that promote the known differential clinical profile in these patient populations.
Collapse
Affiliation(s)
- Hussein Chehade
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan.
- Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan.
| | - Radhika Gogoi
- Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan.
- Karmanos Cancer Institute, Detroit, Michigan.
| | - Nicholas K. Adzibolosu
- Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan.
| | - Sandra Galoforo
- Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan.
| | - Rouba-Ali Fehmi
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan.
- Karmanos Cancer Institute, Detroit, Michigan.
| | - Mira Kheil
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan.
- Karmanos Cancer Institute, Detroit, Michigan.
| | - Alexandra Fox
- Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan.
| | - Seongho Kim
- Karmanos Cancer Institute, Detroit, Michigan.
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.
| | - Ramandeep Rattan
- Division of Gynecology Oncology, Department of Women’s Health Services, Henry Ford Cancer Institute and Henry Ford Health System, Detroit, Michigan.
| | - Zhanjun Hou
- Karmanos Cancer Institute, Detroit, Michigan.
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.
| | - Robert T. Morris
- Karmanos Cancer Institute, Detroit, Michigan.
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.
| | - Larry H. Matherly
- Karmanos Cancer Institute, Detroit, Michigan.
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.
| | - Gil Mor
- Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan.
- Karmanos Cancer Institute, Detroit, Michigan.
| | - Ayesha B. Alvero
- Department of Obstetrics and Gynecology, C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan.
- Karmanos Cancer Institute, Detroit, Michigan.
| |
Collapse
|
7
|
Liu YB, Tan XH, Yang HH, Yang JT, Zhang CY, Jin L, Yang NSY, Guan CX, Zhou Y, Liu SK, Xiong JB. Wnt5a-mediated autophagy contributes to the epithelial-mesenchymal transition of human bronchial epithelial cells during asthma. Mol Med 2024; 30:93. [PMID: 38898476 PMCID: PMC11188189 DOI: 10.1186/s10020-024-00862-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND The epithelial-mesenchymal transition (EMT) of human bronchial epithelial cells (HBECs) is essential for airway remodeling during asthma. Wnt5a has been implicated in various lung diseases, while its role in the EMT of HBECs during asthma is yet to be determined. This study sought to define whether Wnt5a initiated EMT, leading to airway remodeling through the induction of autophagy in HBECs. METHODS Microarray analysis was used to investigate the expression change of WNT5A in asthma patients. In parallel, EMT models were induced using 16HBE cells by exposing them to house dust mites (HDM) or interleukin-4 (IL-4), and then the expression of Wnt5a was observed. Using in vitro gain- and loss-of-function approaches via Wnt5a mimic peptide FOXY5 and Wnt5a inhibitor BOX5, the alterations in the expression of the epithelial marker E-cadherin and the mesenchymal marker protein were observed. Mechanistically, the Ca2+/CaMKII signaling pathway and autophagy were evaluated. An autophagy inhibitor 3-MA was used to examine Wnt5a in the regulation of autophagy during EMT. Furthermore, we used a CaMKII inhibitor KN-93 to determine whether Wnt5a induced autophagy overactivation and EMT via the Ca2+/CaMKII signaling pathway. RESULTS Asthma patients exhibited a significant increase in the gene expression of WNT5A compared to the healthy control. Upon HDM and IL-4 treatments, we observed that Wnt5a gene and protein expression levels were significantly increased in 16HBE cells. Interestingly, Wnt5a mimic peptide FOXY5 significantly inhibited E-cadherin and upregulated α-SMA, Collagen I, and autophagy marker proteins (Beclin1 and LC3-II). Rhodamine-phalloidin staining showed that FOXY5 resulted in a rearrangement of the cytoskeleton and an increase in the quantity of stress fibers in 16HBE cells. Importantly, blocking Wnt5a with BOX5 significantly inhibited autophagy and EMT induced by IL-4 in 16HBE cells. Mechanistically, autophagy inhibitor 3-MA and CaMKII inhibitor KN-93 reduced the EMT of 16HBE cells caused by FOXY5, as well as the increase in stress fibers, cell adhesion, and autophagy. CONCLUSION This study illustrates a new link in the Wnt5a-Ca2+/CaMKII-autophagy axis to triggering airway remodeling. Our findings may provide novel strategies for the treatment of EMT-related diseases.
Collapse
Affiliation(s)
- Yu-Biao Liu
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Xiao-Hua Tan
- Experimental Center of Medical Morphology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Jin-Tong Yang
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Ling Jin
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Nan-Shi-Yu Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, China
- Key Laboratory of General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan, 410078, China
| | - Shao-Kun Liu
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China.
- Clinical Medical Research Center for Pulmonary and Critical Care Medicine in Hunan Province, Changsha, Hunan, 410011, China.
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan, 410011, China.
| | - Jian-Bing Xiong
- Department of Emergency, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China.
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China.
| |
Collapse
|
8
|
Wang Y, Sun Y, Li X, Yu X, Zhang K, Liu J, Tian Q, Zhang H, Du X, Wang S. Progress in the treatment of malignant ascites. Crit Rev Oncol Hematol 2024; 194:104237. [PMID: 38128628 DOI: 10.1016/j.critrevonc.2023.104237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 11/14/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Malignant ascites occurs as a symptom of the terminal stage of cancer, affecting the quality of life through abdominal distension, pain, nausea, anorexia, dyspnea and other symptoms. We describe the current main drug treatments in addition to surgery according to the traditional and new strategies. Traditional treatments were based on anti-tumor chemotherapy and traditional Chinese medicine treatments, as well as diuretics to relieve the patient's symptoms. New treatments mainly involve photothermal therapy, intestinal therapy and targeted immunity. This study emphasizes that both traditional and new therapies have certain advantages and disadvantages, and medication should be adjusted according to different periods of use and different patients. In conclusion, this article reviews the literature to systematically describe the primary treatment modalities for malignant ascites.
Collapse
Affiliation(s)
- Yiqiu Wang
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou, Zhejiang 311121, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yunting Sun
- Hangzhou TCM Hospital Afflitiated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311121, China.
| | - Xinyue Li
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou, Zhejiang 311121, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Xiaoli Yu
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou, Zhejiang 311121, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Keying Zhang
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou, Zhejiang 311121, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jinglei Liu
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou, Zhejiang 311121, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Qingchang Tian
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou, Zhejiang 311121, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Honghua Zhang
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou, Zhejiang 311121, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Xiao Du
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou, Zhejiang 311121, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Shuling Wang
- Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Hangzhou, Zhejiang 311121, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| |
Collapse
|
9
|
Fang Y, Xiao X, Wang J, Dasari S, Pepin D, Nephew KP, Zamarin D, Mitra AK. Cancer associated fibroblasts serve as an ovarian cancer stem cell niche through noncanonical Wnt5a signaling. NPJ Precis Oncol 2024; 8:7. [PMID: 38191909 PMCID: PMC10774407 DOI: 10.1038/s41698-023-00495-5] [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: 03/28/2023] [Accepted: 12/05/2023] [Indexed: 01/10/2024] Open
Abstract
Frequent relapse and chemoresistance cause poor outcome in ovarian cancer (OC) and cancer stem cells (CSCs) are important contributors. While most studies focus exclusively on CSCs, the role of the microenvironment in providing optimal conditions to maintain their tumor-initiating potential remains poorly understood. Cancer associated fibroblasts (CAFs) are a major constituent of the OC tumor microenvironment and we show that CAFs and CSCs are enriched following chemotherapy in patient tumors. CAFs significantly increase OC cell resistance to carboplatin. Using heterotypic CAF-OC cocultures and in vivo limiting dilution assay, we confirm that the CAFs act by enriching the CSC population. CAFs increase the symmetric division of CSCs as well as the dedifferentiation of bulk OC cells into CSCs. The effect of CAFs is limited to OC cells in their immediate neighborhood, which can be prevented by inhibiting Wnt. Analysis of single cell RNA-seq data from OC patients reveal Wnt5a as the highest expressed Wnt in CAFs and that certain subpopulations of CAFs express higher levels of Wnt5a. Our findings demonstrate that Wnt5a from CAFs activate a noncanonical Wnt signaling pathway involving the ROR2/PKC/CREB1 axis in the neighboring CSCs. While canonical Wnt signaling is found to be predominant in interactions between cancer cells in patients, non-canonical Wnt pathway is activated by the CAF-OC crosstalk. Treatment with a Wnt5a inhibitor sensitizes tumors to carboplatin in vivo. Together, our results demonstrate a novel mechanism of CSC maintenance by signals from the microenvironmental CAFs, which can be targeted to treat OC chemoresistance and relapse.
Collapse
Affiliation(s)
- Yiming Fang
- Indiana University School of Medicine-Bloomington, Indiana University, Bloomington, IN, USA
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xue Xiao
- Indiana University School of Medicine-Bloomington, Indiana University, Bloomington, IN, USA
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ji Wang
- Indiana University School of Medicine-Bloomington, Indiana University, Bloomington, IN, USA
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Subramanyam Dasari
- Indiana University School of Medicine-Bloomington, Indiana University, Bloomington, IN, USA
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David Pepin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Kenneth P Nephew
- Indiana University School of Medicine-Bloomington, Indiana University, Bloomington, IN, USA
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Dmitriy Zamarin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anirban K Mitra
- Indiana University School of Medicine-Bloomington, Indiana University, Bloomington, IN, USA.
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
10
|
Chen J, Hu Q, Zhou C, Jin D. CCT2 prevented β-catenin proteasomal degradation to sustain cancer stem cell traits and promote tumor progression in epithelial ovarian cancer. Mol Biol Rep 2024; 51:54. [PMID: 38165547 DOI: 10.1007/s11033-023-09047-3] [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: 03/26/2023] [Accepted: 10/25/2023] [Indexed: 01/04/2024]
Abstract
BACKGROUND Epithelial ovarian cancer (EOC) is featured by rapid progression and dismal outcomes clinically. Chaperonin Containing TCP1 Subunit 2 (CCT2) was identified as a crucial regulator for tumor progression, however, its exact role in EOC remained largely unknown. METHODS CCT2 expression and prognostic value in EOC samples were assessed according to TCGA dataset. Proliferation and mobility potentials were assessed by CCK8, colony-formation, wound healing, and Transwell assays. Cancer stem cell (CSC) traits were evaluated by RT-PCR, WB assays, sphere-forming assay and chemoresistance analysis. Bioinformatic analysis, co-IP assays and ubiquitin assays were performed to explore the mechanisms of CCT2 on EOC cells. RESULTS CCT2 highly expressed in EOC tissues and predicted poor prognosis of EOC patients by TCGA analysis. Silencing CCT2 significantly restrained cell proliferation, migration, and invasion. Moreover, CCT2 could effectively trigger epithelial-mesenchymal transition to confer extensive invasion potentials to EOC cells, Importantly, CCT2 positively correlated with CSC markers in EOC, and CCT2 knockdown impaired CSC traits and sensitize EOC cells to conventional chemotherapy regimens. Contrarily, overexpressing CCT2 achieved opposite results. Mechanistically, CCT2 exerted its pro-oncogene function by triggering Wnt/β-catenin signaling. Specifically, CCT2 could recruit HSP105-PP2A complex, a well-established dephosphorylation complex, to β-catenin via direct physical interaction to prevent phosphorylation-induced proteasomal degradation of β-catenin, resulting in intracellular accumulation of active β-catenin and increased signaling activity. CONCLUSIONS CCT2 was a novel promotor for EOC progression and a crucial sustainer for CSC traits mainly by preventing β-catenin degradation. Targeting CCT2 may represent a promising therapeutic strategy for EOC.
Collapse
Affiliation(s)
- Jiayao Chen
- Department of Laboratory Medicine, Zhoushan Hospital of Zhejiang Province, Zhoushan, 316021, Zhejiang, China.
| | - Qiong Hu
- Department of Laboratory Medicine, Zhoushan Hospital of Zhejiang Province, Zhoushan, 316021, Zhejiang, China
| | - Chenhao Zhou
- Department of Laboratory Medicine, Zhoushan Hospital of Zhejiang Province, Zhoushan, 316021, Zhejiang, China
| | - Danwen Jin
- Pathological Diagnosis Center, Zhoushan Hospital of Zhejiang Province, Zhoushan, 316021, Zhejiang, China
| |
Collapse
|
11
|
Liu H, Zhou L, Cheng H, Wang S, Luan W, Cai E, Ye X, Zhu H, Cui H, Li Y, Chang X. Characterization of candidate factors associated with the metastasis and progression of high-grade serous ovarian cancer. Chin Med J (Engl) 2023; 136:2974-2982. [PMID: 37284741 PMCID: PMC10752471 DOI: 10.1097/cm9.0000000000002328] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND High-grade serous ovarian cancer (HGSOC) is the biggest cause of gynecological cancer-related mortality because of its extremely metastatic nature. This study aimed to explore and evaluate the characteristics of candidate factors associated with the metastasis and progression of HGSOC. METHODS Transcriptomic data of HGSOC patients' samples collected from primary tumors and matched omental metastatic tumors were obtained from three independent studies in the National Center for Biotechnology Information (NCBI) Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were selected to evaluate the effects on the prognosis and progression of ovarian cancer using data from The Cancer Genome Atlas (TCGA) database. Hub genes' immune landscapes were estimated by the Tumor Immune Estimation Resource (TIMER) database. Finally, using 25 HGSOC patients' cancer tissues and 10 normal fallopian tube tissues, immunohistochemistry (IHC) was performed to quantify the expression levels of hub genes associated with International Federation of Gynecology and Obstetrics (FIGO) stages. RESULTS Fourteen DEGs, ADIPOQ , ALPK2 , BARX1 , CD37 , CNR2 , COL5A3 , FABP4 , FAP , GPR68 , ITGBL1 , MOXD1 , PODNL1 , SFRP2 , and TRAF3IP3 , were upregulated in metastatic tumors in every database while CADPS , GATA4 , STAR , and TSPAN8 were downregulated. ALPK2 , FAP , SFRP2 , GATA4 , STAR , and TSPAN8 were selected as hub genes significantly associated with survival and recurrence. All hub genes were correlated with tumor microenvironment infiltration, especially cancer-associated fibroblasts and natural killer (NK) cells. Furthermore, the expression of FAP and SFRP2 was positively correlated with the International Federation of Gynecology and Obstetrics (FIGO) stage, and their increased protein expression levels in metastatic samples compared with primary tumor samples and normal tissues were confirmed by IHC ( P = 0.0002 and P = 0.0001, respectively). CONCLUSIONS This study describes screening for DEGs in HGSOC primary tumors and matched metastasis tumors using integrated bioinformatics analyses. We identified six hub genes that were correlated with the progression of HGSOC, particularly FAP and SFRP2 , which might provide effective targets to predict prognosis and provide novel insights into individual therapeutic strategies for HGSOC.
Collapse
Affiliation(s)
- Huiping Liu
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China
- Center of Gynecologic Oncology, Peking University People's Hospital, Beijing 100044, China
| | - Ling Zhou
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China
- Center of Gynecologic Oncology, Peking University People's Hospital, Beijing 100044, China
| | - Hongyan Cheng
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China
- Center of Gynecologic Oncology, Peking University People's Hospital, Beijing 100044, China
| | - Shang Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China
- Center of Gynecologic Oncology, Peking University People's Hospital, Beijing 100044, China
| | - Wenqing Luan
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China
- Center of Gynecologic Oncology, Peking University People's Hospital, Beijing 100044, China
| | - E Cai
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China
- Center of Gynecologic Oncology, Peking University People's Hospital, Beijing 100044, China
| | - Xue Ye
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China
- Center of Gynecologic Oncology, Peking University People's Hospital, Beijing 100044, China
| | - Honglan Zhu
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China
- Center of Gynecologic Oncology, Peking University People's Hospital, Beijing 100044, China
| | - Heng Cui
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China
- Center of Gynecologic Oncology, Peking University People's Hospital, Beijing 100044, China
| | - Yi Li
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China
- Center of Gynecologic Oncology, Peking University People's Hospital, Beijing 100044, China
| | - Xiaohong Chang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China
- Center of Gynecologic Oncology, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
12
|
Geng Z, Pan X, Xu J, Jia X. Friend and foe: the regulation network of ascites components in ovarian cancer progression. J Cell Commun Signal 2023; 17:391-407. [PMID: 36227507 PMCID: PMC10409702 DOI: 10.1007/s12079-022-00698-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 09/11/2022] [Indexed: 10/17/2022] Open
Abstract
The tumor microenvironment (TME) and its complex role in cancer progression have been hotspots of cancer research in recent years. Ascites, which occurs frequently in patients with ovarian cancer especially in advanced stages, represents a unique TME. Malignant ascites contains abundant cellular and acellular components that play important roles in tumorigenesis, growth, metastasis, and chemoresistance of ovarian cancer through complex molecular mechanisms and signaling pathways. As a valuable liquid biopsy sample, ascites fluid is also of great significance for the prognostic analysis of ovarian cancer. The components of ovarian cancer ascites are generally considered to comprise tumor-promoting factors; however, in recent years studies have found that ascites also contains tumor-suppressing factors, raising new perspectives on interactions between ascites and tumors. Malignant ascites directly constitutes the ovarian cancer microenvironment, therefore, the study of its components will aid in the development of new therapeutic strategies. This article reviews the current research on tumor-promoting and tumor-suppressing factors and molecular mechanisms of their actions in ovarian cancer-derived ascites and therapeutic strategies targeting ascites, which may provide references for the development of novel therapeutic targets for ovarian cancer in the future.
Collapse
Affiliation(s)
- Zhe Geng
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, 123 Mochou Rd, Nanjing, 210004, China
| | - Xinxing Pan
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, 123 Mochou Rd, Nanjing, 210004, China
| | - Juan Xu
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, 123 Mochou Rd, Nanjing, 210004, China.
| | - Xuemei Jia
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, 123 Mochou Rd, Nanjing, 210004, China.
| |
Collapse
|
13
|
Kawamura T, Tanaka N, Hori M, Inoue KI, Kawamura M, Matsusaki K. Hemodynamic Variability During Drainage of Large Volumes of Malignant Ascites in Patients With Cancer. Clin Nurs Res 2023; 32:815-820. [PMID: 36856297 DOI: 10.1177/10547738231157157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
This study compared the variability in mean arterial pressure (MAP) during drainage of ascites in patients with cancer who underwent drainage of a large (5-10 L) or small (<5 L) volume of ascites. We prospectively enrolled 50 patients scheduled for cell-free and concentrated ascites reinfusion therapy. Equivalence was considered to be established if the 95% confidence interval (95% CI) for the highest variability rate of MAP was within ±20%. The median volume of ascites removed was 3.30 L (95% CI [2.84, 4.40]) in the small-drainage-volume group (n = 15) and 6.75 L (95% CI [6.40, 7.30]) in the large-drainage-volume group (n = 34). The 95% CIs for the highest variability rates in MAP ranged from -19.60 to -6.23 and from -19.16 to -12.95 (p = .594), respectively, indicating equivalence. These findings indicate that variability in MAP during drainage of ascites is not dependent on drainage volume.
Collapse
|
14
|
Song Y, Pan S, Li K, Chen X, Wang ZP, Zhu X. Insight into the role of multiple signaling pathways in regulating cancer stem cells of gynecologic cancers. Semin Cancer Biol 2022; 85:219-233. [PMID: 34098106 DOI: 10.1016/j.semcancer.2021.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/24/2021] [Accepted: 06/01/2021] [Indexed: 12/29/2022]
Abstract
Mounting evidence has demonstrated that a myriad of developmental signaling pathways, such as the Wnt, Notch, Hedgehog and Hippo, are frequently deregulated and play a critical role in regulating cancer stem cell (CSC) activity in human cancers, including gynecologic malignancies. In this review article, we describe an overview of various signaling pathways in human cancers. We further discuss the developmental roles how these pathways regulate CSCs from experimental evidences in gynecologic cancers. Moreover, we mention several compounds targeting CSCs in gynecologic cancers to enhance the treatment outcomes. Therefore, these signaling pathways might be the potential targets for developing targeted therapy in gynecologic cancers.
Collapse
Affiliation(s)
- Yizuo Song
- Center of Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Shuya Pan
- Center of Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Kehan Li
- Center of Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Xin Chen
- Center of Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Z Peter Wang
- Center of Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China.
| | - Xueqiong Zhu
- Center of Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang, China.
| |
Collapse
|
15
|
WNT5A in tumor development and progression: A comprehensive review. Biomed Pharmacother 2022; 155:113599. [PMID: 36089446 DOI: 10.1016/j.biopha.2022.113599] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/18/2022] [Accepted: 08/21/2022] [Indexed: 11/20/2022] Open
Abstract
The investigation of tumor microenvironment (TME) is essential to better characterize the complex cellular crosstalk and to identify important immunological phenotypes and biomarkers. The niche is a crucial contributor to neoplasm initiation, maintenance and progression. Therefore, a deeper analysis of tumor surroundings could improve cancer diagnosis, prognosis and assertive treatment. Thus, the WNT family exerts a critical action in tumorigenesis of different types of neoplasms due to dysregulations in the TME. WNT5A, an evolutionary WNT member, is involved in several cellular and physiopathological processes, in addition to tissue homeostasis. The WNT5A protein exerts paradoxical effects while acting as both an oncogene or tumor suppressor by regulating several non-canonical signaling pathways, and consequently interfering in cell growth, cytoskeletal remodeling, migration and invasiveness. This review focuses on a thorough characterization of the role of WNT5A in neoplastic transformation and progression, which may help to understand the prognostic potentiality of WNT5A and its features as a therapeutic target in several cancers. Additionally, we herein summarized novel findings on the mechanisms by which WNT5A might favor tumorigenesis or suppression of cancer progression and discussed the recently developed treatment strategies using WNT5A as a protagonist.
Collapse
|
16
|
Li Y, Wang F, Liu T, Lv N, Yuan X, Li P. WISP1 induces ovarian cancer via the IGF1/αvβ3/Wnt axis. J Ovarian Res 2022; 15:94. [PMID: 35964060 PMCID: PMC9375285 DOI: 10.1186/s13048-022-01016-x] [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: 09/23/2021] [Accepted: 07/04/2022] [Indexed: 11/26/2022] Open
Abstract
Background This study intended to clarify the mechanisms by which WISP1-mediated IGF1/αvβ3/Wnt axis might affect the progression of ovarian cancer. Methods Bioinformatics analysis was implemented for pinpointing expression of IGF1 and WISP1 which was verified through expression determination in clinical tissue samples and cells. Next, gain- or loss-of-function experimentations were implemented for testing CAOV4 and SKOV3 cell biological processes. The interaction between WISP1 and IGF1 was verified by co-immunoprecipitation and the molecular mechanism was analyzed. Finally, ovarian cancer nude mouse models were prepared to unveil the in vivo effects of WISP1/IGF1. Results IGF1 and WISP1 expression was elevated in ovarian cancer tissues and cells, which shared correlation with poor prognosis of ovarian cancer sufferers. Elevated IGF1 induced malignant properties of ovarian cancer cells through activation of PI3K-Akt and Wnt signaling pathway. WISP1 was positively correlated with IGF1. WISP1 could enhance the interaction between IGF1 and αvβ3 to induce epithelial-mesenchymal transition. In vivo experiments also confirmed that upregulated WISP1/IGF1 induced tumorigenesis and metastasis of ovarian cancer cells. Conclusion In conclusion, WISP1 can facilitate ovarian cancer by activating Wnt via the interaction between IGF1 and αvβ3. Supplementary Information The online version contains supplementary material available at 10.1186/s13048-022-01016-x.
Collapse
Affiliation(s)
- Yan Li
- 3th Ward of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, People's Republic of China
| | - Fangfang Wang
- Prenatal Diagnosis Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, People's Republic of China
| | - Tianyi Liu
- 3th Ward of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, People's Republic of China
| | - Nan Lv
- 3th Ward of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, People's Republic of China
| | - Xiaolei Yuan
- 3th Ward of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, People's Republic of China
| | - Peiling Li
- 1st Ward of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, No. 246, Xuefu Road, Nangang District, Harbin, 150086, People's Republic of China.
| |
Collapse
|
17
|
The Cell Surface Heparan Sulfate Proteoglycan Syndecan-3 Promotes Ovarian Cancer Pathogenesis. Int J Mol Sci 2022; 23:ijms23105793. [PMID: 35628603 PMCID: PMC9145288 DOI: 10.3390/ijms23105793] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 11/24/2022] Open
Abstract
Syndecans are transmembrane heparan sulfate proteoglycans that integrate signaling at the cell surface. By interacting with cytokines, signaling receptors, proteases, and extracellular matrix proteins, syndecans regulate cell proliferation, metastasis, angiogenesis, and inflammation. We analyzed public gene expression datasets to evaluate the dysregulation and potential prognostic impact of Syndecan-3 in ovarian cancer. Moreover, we performed functional in vitro analysis in syndecan-3-siRNA-treated SKOV3 and CAOV3 ovarian cancer cells. In silico analysis of public gene array datasets revealed that syndecan-3 mRNA expression was significantly increased 5.8-fold in ovarian cancer tissues (n = 744) and 3.4-fold in metastases (n = 44) compared with control tissue (n = 46), as independently confirmed in an RNAseq dataset on ovarian serous cystadenocarcinoma tissue (n = 374, controls: n = 133, 3.5-fold increase tumor vs. normal). Syndecan-3 siRNA knockdown impaired 3D spheroid growth and colony formation as stemness-related readouts in SKOV3 and CAOV3 cells. In SKOV3, but not in CAOV3 cells, syndecan-3 depletion reduced cell viability both under basal conditions and under chemotherapy with cisplatin, or cisplatin and paclitaxel. While analysis of the SIOVDB database did not reveal differences in Syndecan-3 expression between patients, sensitive, resistant or refractory to chemotherapy, KM Plotter analysis of 1435 ovarian cancer patients revealed that high syndecan-3 expression was associated with reduced survival in patients treated with taxol and platin. At the molecular level, a reduction in Stat3 activation and changes in the expression of Wnt and notch signaling constituents were observed. Our study suggests that up-regulation of syndecan-3 promotes the pathogenesis of ovarian cancer by modulating stemness-associated pathways.
Collapse
|
18
|
Shepherd TG, Dick FA. Principles of dormancy evident in high-grade serous ovarian cancer. Cell Div 2022; 17:2. [PMID: 35321751 PMCID: PMC8944075 DOI: 10.1186/s13008-022-00079-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 03/16/2022] [Indexed: 11/17/2022] Open
Abstract
In cancer, dormancy refers to a clinical state in which microscopic residual disease becomes non-proliferative and is largely refractory to chemotherapy. Dormancy was first described in breast cancer where disease can remain undetected for decades, ultimately leading to relapse and clinical presentation of the original malignancy. A long latency period can be explained by withdrawal from cell proliferation (cellular dormancy), or a balance between proliferation and cell death that retains low levels of residual disease (tumor mass dormancy). Research into cellular dormancy has revealed features that define this state. They include arrest of cell proliferation, altered cellular metabolism, and unique cell dependencies and interactions with the microenvironment. These characteristics can be shared by dormant cells derived from disparate primary disease sites, suggesting common features exist between them. High-grade serous ovarian cancer (HGSOC) disseminates to locations throughout the abdominal cavity by means of cellular aggregates called spheroids. These growth-arrested and therapy-resistant cells are a strong contributor to disease relapse. In this review, we discuss the similarities and differences between ovarian cancer cells in spheroids and dormant properties reported for other cancer disease sites. This reveals that elements of dormancy, such as cell cycle control mechanisms and changes to metabolism, may be similar across most forms of cellular dormancy. However, HGSOC-specific aspects of spheroid biology, including the extracellular matrix organization and microenvironment, are obligatorily disease site specific. Collectively, our critical review of current literature highlights places where HGSOC cell dormancy may offer a more tractable experimental approach to understand broad principles of cellular dormancy in cancer.
Collapse
Affiliation(s)
- Trevor G Shepherd
- London Regional Cancer Program, London Health Sciences Centre, London, ON, N6A 5W9, Canada.,Department of Obstetrics & Gynaecology, Western University, London, ON, N6A 5C1, Canada
| | - Frederick A Dick
- London Regional Cancer Program, London Health Sciences Centre, London, ON, N6A 5W9, Canada. .,Department of Pathology and Laboratory Medicine, Western University, London, ON, N6A 5C1, Canada. .,Children's Health Research Institute, London, ON, N6A 4V2, Canada.
| |
Collapse
|
19
|
Iyoshi S, Yoshihara M, Nakamura K, Sugiyama M, Koya Y, Kitami K, Uno K, Mogi K, Tano S, Tomita H, Kajiwara K, Taki M, Yamaguchi S, Nawa A, Kajiyama H. Pro-tumoral behavior of omental adipocyte-derived fibroblasts in tumor microenvironment at the metastatic site of ovarian cancer. Int J Cancer 2021; 149:1961-1972. [PMID: 34469585 DOI: 10.1002/ijc.33770] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 07/08/2021] [Accepted: 08/06/2021] [Indexed: 12/16/2022]
Abstract
Adipocyte-rich omentum offers "good soil" for disseminating ovarian cancer (OvCa), contributing to therapeutic difficulty. However, little is understood about the association between adipocytes and tumor growth at peritoneal dissemination site. Herein, we report the induction of adipocyte dedifferentiation by OvCa cells and pro-tumorigenic effects of resulted adipocyte-derived fibroblasts. We confirmed that malignant ascites promoted the dedifferentiation of the primary human adipocytes obtained from surgical omental specimen into omental adipocyte-derived fibroblast (O-ADF) that possess both mesenchymal stem cell and myofibroblast-like features. This promotion of dedifferentiation by malignant ascites was blocked by addition of Wnt signaling inhibitor. The effects of dedifferentiated adipocytes in proliferation and migration of OvCa cells were analyzed with in vitro coculturing experimental models and in vivo mice model, and we demonstrated that OvCa cell lines showed enhanced proliferative characteristics, as well as increased migratory abilities upon coculturing with O-ADF. Additionally, exogenous transforming growth factor-β1 augmented desmoplastic morphological change of O-ADF, leading to higher proliferative ability. Our results suggest that OvCa cells promote dedifferentiation of peritoneal adipocytes by activating Wnt/β-catenin signaling, and generated O-ADFs exhibit pro-tumoral hallmarks.
Collapse
Affiliation(s)
- Shohei Iyoshi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Masato Yoshihara
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kae Nakamura
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Center for Low-Temperature Plasma Sciences, Nagoya University, Nagoya, Japan
| | - Mai Sugiyama
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihiro Koya
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuhisa Kitami
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kaname Uno
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Division of Clinical Genetics, Department of Laboratory Medicine, Lund University Graduate School of Medicine, Lund, Sweden
| | - Kazumasa Mogi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sho Tano
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Keiji Kajiwara
- Department of Chemistry, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Masayasu Taki
- Institute of Transformative Bio-Molecules, Nagoya University, Nagoya, Japan
| | - Shigehiro Yamaguchi
- Department of Chemistry, Graduate School of Science, Nagoya University, Nagoya, Japan.,Institute of Transformative Bio-Molecules, Nagoya University, Nagoya, Japan
| | - Akihiro Nawa
- Bell Research Center, Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
20
|
Sommerfeld L, Finkernagel F, Jansen JM, Wagner U, Nist A, Stiewe T, Müller‐Brüsselbach S, Sokol AM, Graumann J, Reinartz S, Müller R. The multicellular signalling network of ovarian cancer metastases. Clin Transl Med 2021; 11:e633. [PMID: 34841720 PMCID: PMC8574964 DOI: 10.1002/ctm2.633] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/08/2021] [Accepted: 10/15/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Transcoelomic spread is the major route of metastasis of ovarian high-grade serous carcinoma (HGSC) with the omentum as the major metastatic site. Its unique tumour microenvironment with its large populations of adipocytes, mesothelial cells and immune cells establishes an intercellular signaling network that is instrumental for metastatic growth yet poorly understood. METHODS Based on transcriptomic analysis of tumour cells, tumour-associated immune and stroma cells we defined intercellular signaling pathways for 284 cytokines and growth factors and their cognate receptors after bioinformatic adjustment for contaminating cell types. The significance of individual components of this network was validated by analysing clinical correlations and potentially pro-metastatic functions, including tumour cell migration, pro-inflammatory signal transduction and TAM expansion. RESULTS The data show an unexpected prominent role of host cells, and in particular of omental adipocytes, mesothelial cells and fibroblasts (CAF), in sustaining this signaling network. These cells, rather than tumour cells, are the major source of most cytokines and growth factors in the omental microenvironment (n = 176 vs. n = 13). Many of these factors target tumour cells, are linked to metastasis and are associated with a short survival. Likewise, tumour stroma cells play a major role in extracellular-matrix-triggered signaling. We have verified the functional significance of our observations for three exemplary instances. We show that the omental microenvironment (i) stimulates tumour cell migration and adhesion via WNT4 which is highly expressed by CAF; (ii) induces pro-tumourigenic TAM proliferation in conjunction with high CSF1 expression by omental stroma cells and (iii) triggers pro-inflammatory signaling, at least in part via a HSP70-NF-κB pathway. CONCLUSIONS The intercellular signaling network of omental metastases is majorly dependent on factors secreted by immune and stroma cells to provide an environment that supports ovarian HGSC progression. Clinically relevant pathways within this network represent novel options for therapeutic intervention.
Collapse
Affiliation(s)
- Leah Sommerfeld
- Department of Translational Oncology, Center for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Florian Finkernagel
- Department of Translational Oncology, Center for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Julia M. Jansen
- Clinic for Gynecology, Gynecological Oncology and Gynecological EndocrinologyUniversity Hospital (UKGM)MarburgGermany
| | - Uwe Wagner
- Clinic for Gynecology, Gynecological Oncology and Gynecological EndocrinologyUniversity Hospital (UKGM)MarburgGermany
| | - Andrea Nist
- Genomics Core Facility, Center for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Thorsten Stiewe
- Genomics Core Facility, Center for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
- Institute of Molecular OncologyPhilipps UniversityMarburgGermany
| | - Sabine Müller‐Brüsselbach
- Department of Translational Oncology, Center for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Anna M. Sokol
- The German Centre for Cardiovascular Research (DZHK), Partner Site Rhine‐MainMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Johannes Graumann
- The German Centre for Cardiovascular Research (DZHK), Partner Site Rhine‐MainMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
- Institute for Translational Proteomics, Philipps UniversityMarburgGermany
| | - Silke Reinartz
- Department of Translational Oncology, Center for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| | - Rolf Müller
- Department of Translational Oncology, Center for Tumor Biology and Immunology (ZTI)Philipps UniversityMarburgGermany
| |
Collapse
|
21
|
Radaszkiewicz T, Nosková M, Gömöryová K, Vondálová Blanářová O, Radaszkiewicz KA, Picková M, Víchová R, Gybeľ T, Kaiser K, Demková L, Kučerová L, Bárta T, Potěšil D, Zdráhal Z, Souček K, Bryja V. RNF43 inhibits WNT5A-driven signaling and suppresses melanoma invasion and resistance to the targeted therapy. eLife 2021; 10:65759. [PMID: 34702444 PMCID: PMC8550759 DOI: 10.7554/elife.65759] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 09/28/2021] [Indexed: 12/30/2022] Open
Abstract
RNF43 is an E3 ubiquitin ligase and known negative regulator of WNT/β-catenin signaling. We demonstrate that RNF43 is also a regulator of noncanonical WNT5A-induced signaling in human cells. Analysis of the RNF43 interactome using BioID and immunoprecipitation showed that RNF43 can interact with the core receptor complex components dedicated to the noncanonical Wnt pathway such as ROR1, ROR2, VANGL1, and VANGL2. RNF43 triggers VANGL2 ubiquitination and proteasomal degradation and clathrin-dependent internalization of ROR1 receptor and inhibits ROR2 activation. These activities of RNF43 are physiologically relevant and block pro-metastatic WNT5A signaling in melanoma. RNF43 inhibits responses to WNT5A, which results in the suppression of invasive properties of melanoma cells. Furthermore, RNF43 prevented WNT5A-assisted development of resistance to BRAF V600E and MEK inhibitors. Next, RNF43 acted as melanoma suppressor and improved response to targeted therapies in vivo. In line with these findings, RNF43 expression decreases during melanoma progression and RNF43-low patients have a worse prognosis. We conclude that RNF43 is a newly discovered negative regulator of WNT5A-mediated biological responses that desensitizes cells to WNT5A.
Collapse
Affiliation(s)
- Tomasz Radaszkiewicz
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Michaela Nosková
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Kristína Gömöryová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Olga Vondálová Blanářová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | | | - Markéta Picková
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.,Department of Cytokinetics, Institute of Biophysics CAS, Brno, Czech Republic.,International Clinical Research Center FNUSA-ICRC, Brno, Czech Republic
| | - Ráchel Víchová
- Department of Cytokinetics, Institute of Biophysics CAS, Brno, Czech Republic
| | - Tomáš Gybeľ
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Karol Kaiser
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Lucia Demková
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lucia Kučerová
- Laboratory of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Tomáš Bárta
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.,Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - David Potěšil
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Zbyněk Zdráhal
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Karel Souček
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.,Department of Cytokinetics, Institute of Biophysics CAS, Brno, Czech Republic.,International Clinical Research Center FNUSA-ICRC, Brno, Czech Republic
| | - Vítězslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.,Department of Cytokinetics, Institute of Biophysics CAS, Brno, Czech Republic
| |
Collapse
|
22
|
Qian C, Liu Q. FOXO3a inhibits nephroblastoma cell proliferation, migration and invasion, and induces apoptosis through downregulating the Wnt/β‑catenin signaling pathway. Mol Med Rep 2021; 24:796. [PMID: 34515328 PMCID: PMC8446726 DOI: 10.3892/mmr.2021.12436] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 06/26/2020] [Indexed: 12/18/2022] Open
Abstract
Forkhead transcription factor O subfamily 3A (FOXO3a) is an important tumor suppressor gene that is expressed in renal tissue and has been reported to be downregulated in clear cell renal cell carcinoma (CCRCC). Notably, the overexpression of FOXO3a was previously discovered to inhibit the progression of CCRCC. However, the expression levels of FOXO3a in nephroblastoma cell lines remain unknown. The present study aimed to investigate the expression levels of FOXO3a in nephroblastoma cell lines and to determine the mechanism of action of the biological functions of FOXO3a. Western blotting and reverse transcription‑quantitative PCR were used to analyze the expression levels of FOXO3a in nephroblastoma cell lines. Subsequently, the effects of the overexpression of FOXO3a and the genetic knockdown of the Wnt/β‑catenin signaling protein Axin‑2 on the biological functions were determined through Cell Counting Kit‑8, cell colony formation assays, scratch and Transwell assay and flow cytometric analysis experiments. The expression levels of FOXO3a were discovered to be downregulated in nephroblastoma cell lines. The overexpression of FOXO3a inhibited the proliferation, invasion and migration of nephroblastoma cells, while inducing apoptosis. Furthermore, the overexpression of FOXO3a downregulated the expression levels of β‑catenin and Cyclin‑D1 proteins involved in the Wnt/β‑catenin signaling pathway. Cell proliferation and the migration and invasion ability of 17‑94 cells in shRNA‑Axin2‑2 group were promoted. Cell apoptosis was predominantly increased by overexpressed FOXO3a, which was reversed by shRNA‑Axin2‑1. The biological effects of overexpressing FOXO3a on nephroblastoma were reversed after activation of Wnt/β‑catenin. In conclusion, the findings of the present study suggested that FOXO3a may inhibit nephroblastoma cell proliferation, migration and invasion, while inducing apoptosis, by downregulating the Wnt/β‑catenin signaling pathway. These results may provide a novel method for the early diagnosis and precise treatment of nephroblastoma.
Collapse
Affiliation(s)
- Cheng Qian
- Department of Pediatric Surgery, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277102, P.R. China
| | - Qiang Liu
- Department of Urinary Surgery, The Second People's Hospital of Nantong, Nantong, Jiangsu 226002, P.R. China
| |
Collapse
|
23
|
Záveský L, Jandáková E, Weinberger V, Hanzíková V, Slanař O, Kohoutová M. Ascites in ovarian cancer: MicroRNA deregulations and their potential roles in ovarian carcinogenesis. Cancer Biomark 2021; 33:1-16. [PMID: 34511487 DOI: 10.3233/cbm-210219] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ovarian cancer comprises the most lethal gynecologic malignancy and is accompanied by the high potential for the incidence of metastasis, recurrence and chemotherapy resistance, often associated with a formation of ascitic fluid. The differentially expressed ascites-derived microRNAs may be linked to ovarian carcinogenesis. The article focuses on a number of miRNAs that share a common expression pattern as determined by independent studies using ascites samples and with regard to their functions and outcomes in experimental and clinical investigations.Let-7b and miR-143 have featured as tumor suppressors in ovarian cancer, which is in line with data on other types of cancer. Although two miRNAs, i.e. miR-26a-5p and miR-145-5p, act principally as tumor suppressor miRNAs, they occasionally exhibit oncogenic roles. The performance of miR-95-3p, upregulated in ascites, is open to debate given the current lack of supportive data on ovarian cancer; however, data on other cancers indicates its probable oncogenic role. Different findings have been reported for miR-182-5p and miR-200c-3p; in addition to their presumed oncogenic roles, contrasting findings have indicated their ambivalent functions. Further research is required for the identification and evaluation of the potential of specific miRNAs in the diagnosis, prediction, treatment and outcomes of ovarian cancer patients.
Collapse
Affiliation(s)
- Luděk Záveský
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague and General University Hospital, Prague, Czech Republic.,Institute of Pharmacology, First Faculty of Medicine, Charles University, Prague and General University Hospital, Prague, Czech Republic
| | - Eva Jandáková
- Department of Pathology, Faculty of Medicine, Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Vít Weinberger
- Department of Obstetrics and Gynecology, Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Veronika Hanzíková
- Faculty Transfusion Center, General University Hospital, Prague, Czech Republic
| | - Ondřej Slanař
- Institute of Pharmacology, First Faculty of Medicine, Charles University, Prague and General University Hospital, Prague, Czech Republic
| | - Milada Kohoutová
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague and General University Hospital, Prague, Czech Republic
| |
Collapse
|
24
|
Chan DW, Lam WY, Chen F, Yung MMH, Chan YS, Chan WS, He F, Liu SS, Chan KKL, Li B, Ngan HYS. Genome-wide DNA methylome analysis identifies methylation signatures associated with survival and drug resistance of ovarian cancers. Clin Epigenetics 2021; 13:142. [PMID: 34294135 PMCID: PMC8296615 DOI: 10.1186/s13148-021-01130-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/12/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND In contrast to stable genetic events, epigenetic changes are highly plastic and play crucial roles in tumor evolution and development. Epithelial ovarian cancer (EOC) is a highly heterogeneous disease that is generally associated with poor prognosis and treatment failure. Profiling epigenome-wide DNA methylation status is therefore essential to better characterize the impact of epigenetic alterations on the heterogeneity of EOC. METHODS An epigenome-wide association study was conducted to evaluate global DNA methylation in a retrospective cohort of 80 mixed subtypes of primary ovarian cancers and 30 patients with high-grade serous ovarian carcinoma (HGSOC). Three demethylating agents, azacytidine, decitabine, and thioguanine, were tested their anti-cancer and anti-chemoresistant effects on HGSOC cells. RESULTS Global DNA hypermethylation was significantly associated with high-grade tumors, platinum resistance, and poor prognosis. We determined that 9313 differentially methylated probes (DMPs) were enriched in their relative gene regions of 4938 genes involved in small GTPases and were significantly correlated with the PI3K-AKT, MAPK, RAS, and WNT oncogenic pathways. On the other hand, global DNA hypermethylation was preferentially associated with recurrent HGSOC. A total of 2969 DMPs corresponding to 1471 genes were involved in olfactory transduction, and calcium and cAMP signaling. Co-treatment with demethylating agents showed significant growth retardation in ovarian cancer cells through differential inductions, such as cell apoptosis by azacytidine or G2/M cell cycle arrest by decitabine and thioguanine. Notably, azacytidine and decitabine, though not thioguanine, synergistically enhanced cisplatin-mediated cytotoxicity in HGSOC cells. CONCLUSIONS This study demonstrates the significant association of global hypermethylation with poor prognosis and drug resistance in high-grade EOC and highlights the potential of demethylating agents in cancer treatment.
Collapse
Affiliation(s)
- David W Chan
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam, Hong Kong, SAR, People's Republic of China.
| | - Wai-Yip Lam
- Lee's Pharmaceutical (HK) Ltd, 1/F Building 20E, Phase 3, Hong Kong Science Park, Shatin, Hong Kong, People's Republic of China
| | - Fushun Chen
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam, Hong Kong, SAR, People's Republic of China
| | - Mingo M H Yung
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam, Hong Kong, SAR, People's Republic of China
| | - Yau-Sang Chan
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam, Hong Kong, SAR, People's Republic of China
| | - Wai-Sun Chan
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam, Hong Kong, SAR, People's Republic of China
| | - Fangfang He
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam, Hong Kong, SAR, People's Republic of China
| | - Stephanie S Liu
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam, Hong Kong, SAR, People's Republic of China
| | - Karen K L Chan
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam, Hong Kong, SAR, People's Republic of China
| | - Benjamin Li
- Lee's Pharmaceutical (HK) Ltd, 1/F Building 20E, Phase 3, Hong Kong Science Park, Shatin, Hong Kong, People's Republic of China
| | - Hextan Y S Ngan
- Department of Obstetrics and Gynaecology, L747 Laboratory Block, LKS Faculty of Medicine, 21 Sassoon Road, Pokfulam, Hong Kong, SAR, People's Republic of China. .,Department of Obstetrics and Gynaecology, 6/F Professorial Block, Queen Mary Hospital, Pokfulam, Hong Kong, People's Republic of China.
| |
Collapse
|
25
|
ZEB2 facilitates peritoneal metastasis by regulating the invasiveness and tumorigenesis of cancer stem-like cells in high-grade serous ovarian cancers. Oncogene 2021; 40:5131-5141. [PMID: 34211089 PMCID: PMC8363099 DOI: 10.1038/s41388-021-01913-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/25/2022]
Abstract
Peritoneal metastasis is a common issue in the progression of high-grade serous ovarian cancers (HGSOCs), yet the underlying mechanism remains unconfirmed. We demonstrated that ZEB2, the transcription factor of epithelial–mesenchymal transition (EMT), was upregulated in ascites cells from HGSOC patients and in CD133+ cancer stem-like cells (CSLCs) from epithelial ovarian cancer (EOC) cell lines. SiRNA-mediated knockdown of ZEB2 in EOC cells decreased the percentage of CSLCs and reduced the colony forming potential, cell invasion capacity and expression of pluripotent genes Oct4 and Nanog. Inhibition of ZEB2 also induced cellular apoptosis and impacted the tumorigenicity of ovarian CSLCs. The mesenchymal markers N-cadherin and vimentin were downregulated, while the epithelial marker E-cadherin was upregulated after ZEB2 knockdown. MiR-200a, a molecule that downregulates ZEB2, had the opposite effect of ZEB2 expression in EOC-CSLCs. A retrospective study of 98 HGSOC patients on the relationship of ascites volume, pelvic and abdominal metastasis, International Federation of Gynecology and Obstetrics (FIGO) stage and the malignant involvement of abdominal organs and lymph nodes was performed. Patients with high expression of ZEB2 in tumour tissues had a higher metastasis rate and a poorer prognosis than those with low expression. The parameters of ZEB2 expression and ascites volume were strongly linked with the prognostic outcome of HGSOC patients and had higher hazard ratios. These findings illustrated that ZEB2 facilitates the invasive metastasis of EOC-CSLCs and can predict peritoneal metastasis and a poor prognosis in HGSOC patients.
Collapse
|
26
|
王 卉, 陈 学, 陈 运, 曹 颖, 陈 瑶, 刘 国, 黄 莉. [ENTPD5 gene is highly expressed in epithelial ovarian cancer: analysis based on Oncomine database and bioinformatics]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:555-561. [PMID: 33963715 PMCID: PMC8110460 DOI: 10.12122/j.issn.1673-4254.2021.04.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To investigate the expression of ENTPD5 in epithelial ovarian cancer and explore its clinical implications. OBJECTIVE The expression level of ENTPD5 in epithelial ovarian cancer was analyzed based on data from Oncomine and TCGA databases. The relationship between the expression level of ENTPD5 and clinical characteristics of the patients was analyzed using UALCAN database. Gene enrichment analysis (GSEA) was performed to explore the possible role of ENTPD5 in the occurrence and progression of epithelial ovarian cancer. CIBERSORT package was used to analyze the relationship between the expression of ENTPD5 and immune infiltration. The expression patterns of ENTPD5 were verified in 23 epithelial ovarian cancer tissues and 15 normal ovarian tissues using RT-qPCR and Western blotting; the expression of ENTPD5 protein was also detected immunohistochemically in 50 paraffin-embedded samples of epithelial ovarian cancer and 6 normal ovarian tissues. OBJECTIVE Analysis of Oncomine and TCGA databases showed that the expression of ENTPD5 was significantly higher in epithelial ovarian cancer tissues than in normal ovarian tissues (P < 0.05), and its expression level was negatively correlated with the survival rate of the patients (P < 0.05). Data from UALCAN database showed that the expression level of ENTPD5 was related with the age of patients. The results of GSEA suggested that ENTPD5 was involved in ABC transporter, WNT signaling pathway and insulin signaling, and the expression of ENTPD5 was negatively correlated with the contents of NK cells, mast cells and eosinophils (P < 0.05). In clinical samples of epithelial ovarian cancer tissues, the expression of ENTPD5 was significantly higher than that in normal ovarian tissues at both the mRNA (P < 0.01) and protein (P < 0.05) levels. The paraffinembedded samples also showed significantly higher expressions of ENTPD5 in epithelial ovarian cancer than in normal ovarian tissues (P < 0.05). OBJECTIVE ENTPD5 is highly expressed in epithelial ovarian cancer, which may promote the occurrence and progression of epithelial ovarian cancer by participating in multiple functional processes and cellular immune infiltration.
Collapse
Affiliation(s)
- 卉 王
- 南方医科大学第一临床医学院,广东 广州 510515The first clinical college of Southern Medical University, Guangzhou 510515, China
| | - 学平 陈
- 南方医科大学第一临床医学院,广东 广州 510515The first clinical college of Southern Medical University, Guangzhou 510515, China
| | - 运 陈
- 南方医科大学第一临床医学院,广东 广州 510515The first clinical college of Southern Medical University, Guangzhou 510515, China
| | - 颖诗 曹
- 南方医科大学第一临床医学院,广东 广州 510515The first clinical college of Southern Medical University, Guangzhou 510515, China
| | - 瑶 陈
- 南方医科大学第一临床医学院,广东 广州 510515The first clinical college of Southern Medical University, Guangzhou 510515, China
| | - 国炳 刘
- 南方医科大学南方医院妇产科,广东 广州 510515Southern Medical University of Nanfang Hospital, Obstetrics and Gynecology Department, Guangzhou 510515, China
| | - 莉萍 黄
- 南方医科大学南方医院妇产科,广东 广州 510515Southern Medical University of Nanfang Hospital, Obstetrics and Gynecology Department, Guangzhou 510515, China
| |
Collapse
|
27
|
Zhou W, Mei J, Gu D, Xu J, Wang R, Wang H, Liu C. Wnt5a: A promising therapeutic target in ovarian cancer. Pathol Res Pract 2021; 219:153348. [DOI: 10.1016/j.prp.2021.153348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 02/07/2023]
|
28
|
Azbazdar Y, Karabicici M, Erdal E, Ozhan G. Regulation of Wnt Signaling Pathways at the Plasma Membrane and Their Misregulation in Cancer. Front Cell Dev Biol 2021; 9:631623. [PMID: 33585487 PMCID: PMC7873896 DOI: 10.3389/fcell.2021.631623] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/04/2021] [Indexed: 12/24/2022] Open
Abstract
Wnt signaling is one of the key signaling pathways that govern numerous physiological activities such as growth, differentiation and migration during development and homeostasis. As pathway misregulation has been extensively linked to pathological processes including malignant tumors, a thorough understanding of pathway regulation is essential for development of effective therapeutic approaches. A prominent feature of cancer cells is that they significantly differ from healthy cells with respect to their plasma membrane composition and lipid organization. Here, we review the key role of membrane composition and lipid order in activation of Wnt signaling pathway by tightly regulating formation and interactions of the Wnt-receptor complex. We also discuss in detail how plasma membrane components, in particular the ligands, (co)receptors and extracellular or membrane-bound modulators, of Wnt pathways are affected in lung, colorectal, liver and breast cancers that have been associated with abnormal activation of Wnt signaling. Wnt-receptor complex components and their modulators are frequently misexpressed in these cancers and this appears to correlate with metastasis and cancer progression. Thus, composition and organization of the plasma membrane can be exploited to develop new anticancer drugs that are targeted in a highly specific manner to the Wnt-receptor complex, rendering a more effective therapeutic outcome possible.
Collapse
Affiliation(s)
- Yagmur Azbazdar
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, İzmir, Turkey.,Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, İzmir, Turkey
| | - Mustafa Karabicici
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, İzmir, Turkey.,Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, İzmir, Turkey
| | - Esra Erdal
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, İzmir, Turkey.,Department of Medical Biology and Genetics, Faculty of Medicine, Dokuz Eylul University, İzmir, Turkey
| | - Gunes Ozhan
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, İzmir, Turkey.,Izmir International Biomedicine and Genome Institute (IBG-Izmir), Dokuz Eylul University, İzmir, Turkey
| |
Collapse
|
29
|
Alshareef A, Peters AC, Gélébart P, Chen W, Lai R. Gene Methylation and Silencing of WIF1 Is a Frequent Genetic Abnormality in Mantle Cell Lymphoma. Int J Mol Sci 2021; 22:ijms22020893. [PMID: 33477402 PMCID: PMC7830226 DOI: 10.3390/ijms22020893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/09/2021] [Accepted: 01/12/2021] [Indexed: 11/29/2022] Open
Abstract
We have previously shown that the Wnt canonical pathway (WCP) is constitutively active in most cases of mantle cell lymphoma (MCL). Here, we aimed to elucidate the mechanisms underlying this biochemical deregulation. We hypothesized that gene methylation/silencing of WIF1 (Wnt inhibitory factor-1), a physiologic inhibitor of WCP, contributes to the deregulation of WCP and promotes cell growth in MCL. In support of this hypothesis, we found that the expression of WIF1 was detectable in none of the 4 MCL cell lines, and in only 2 of 5 tumors (40%) examined. Using methylation-specific PCR, we found evidence of gene methylation of WIF1 in 4 of 5 cell lines (80%) and in 24 of 29 (82%) tumors. The addition of the demethylation agent 5-aza-2′-deoxycytidine to Mino and JeKo-1, two WIF1-negative cell lines, restored the expression of WIF1 mRNA in these cells. Gene transfection of WIF1 into JeKo-1 and Mino cells significantly reduced cell growth, and this finding correlated with substantial downregulations of various proteins in WCP, such as β-catenin and pGSK-3β. In conclusion, our results support the concept that gene methylation/silencing of WIF1 is a frequent event in MCL, and this abnormality contributes to the aberrant activation of WCP. These results have provided further evidence that aberrant Wnt signaling is pathogenetically important in MCL and it may represent a potential therapeutic target.
Collapse
Affiliation(s)
- Abdulraheem Alshareef
- Medical Laboratories Technology Department, College of Applied Medical Sciences, Taibah University, Madinah, P.O. Box 41477, Saudi Arabia;
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2E1, Canada; (P.G.); (W.C.)
| | - Anthea C. Peters
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2E1, Canada;
| | - Pascal Gélébart
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2E1, Canada; (P.G.); (W.C.)
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| | - Will Chen
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2E1, Canada; (P.G.); (W.C.)
| | - Raymond Lai
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2E1, Canada; (P.G.); (W.C.)
- Department of Oncology, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Correspondence:
| |
Collapse
|
30
|
Wang J, Feng D, Gao B. An Overview of Potential Therapeutic Agents Targeting WNT/PCP Signaling. Handb Exp Pharmacol 2021; 269:175-213. [PMID: 34463852 DOI: 10.1007/164_2021_533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Since the discovery of the proto-oncogene Wnt1 (Int1) in 1982, WNT signaling has been identified as one of the most important pathways that regulates a wide range of fundamental developmental and physiological processes in multicellular organisms. The canonical WNT signaling pathway depends on the stabilization and translocation of β-catenin and plays important roles in development and homeostasis. The WNT/planar cell polarity (WNT/PCP) signaling, also known as one of the β-catenin-independent WNT pathways, conveys directional information to coordinate polarized cell behaviors. Similar to WNT/β-catenin signaling, disruption or aberrant activation of WNT/PCP signaling also underlies a variety of developmental defects and cancers. However, the pharmacological targeting of WNT/PCP signaling for therapeutic purposes remains largely unexplored. In this review, we briefly discuss WNT/PCP signaling in development and disease and summarize the known drugs/inhibitors targeting this pathway.
Collapse
Affiliation(s)
- Jin Wang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Di Feng
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Bo Gao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.
| |
Collapse
|
31
|
Sun Y, Wang W, Zhao C. Frizzled Receptors in Tumors, Focusing on Signaling, Roles, Modulation Mechanisms, and Targeted Therapies. Oncol Res 2020; 28:661-674. [PMID: 32998794 PMCID: PMC7962935 DOI: 10.3727/096504020x16014648664459] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Wnt molecules play crucial roles in development and adult homeostasis through their receptors Frizzled proteins (Fzds). Fzds mediate canonical β-catenin pathway and various noncanonical β-catenin-independent pathways. Aberrant Fzd signaling is involved in many diseases including cancer. Wnt/β-catenin is a well-established oncogenic pathway involved in almost every aspect of tumor development. However, Fzd-mediated noncanonical Wnt pathways function as both tumor promoters and tumor suppressors depending on cellular context. Fzd-targeted therapies have proven to be effective on cultured tumor cells, tumor cell xenografts, mouse tumor models, and patient-derived xenografts (PDX). Moreover, Fzd-targeted therapies synergize with chemotherapy in preclinical models. However, the occurrence of fragility fractures in patients treated with Fzd-targeted agents such as OMP-54F28 and OMP-18R5 limits the development of this combination. Along with new insights on signaling, roles, and modulation mechanisms of Fzds in human tumors, more Fzd-related therapeutic targets will be developed.
Collapse
Affiliation(s)
- Yu Sun
- Department of Pathophysiology, College of Basic Medical Science, China Medical UniversityShenyangP.R. China
| | - Wei Wang
- Department of Pathophysiology, College of Basic Medical Science, China Medical UniversityShenyangP.R. China
| | - Chenghai Zhao
- Department of Pathophysiology, College of Basic Medical Science, China Medical UniversityShenyangP.R. China
| |
Collapse
|
32
|
Wang Y, Dou L, Qin Y, Yang H, Yan P. OIP5-AS1 contributes to tumorigenesis in hepatocellular carcinoma by miR-300/YY1-activated WNT pathway. Cancer Cell Int 2020; 20:440. [PMID: 32943988 PMCID: PMC7487829 DOI: 10.1186/s12935-020-01467-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 07/31/2020] [Indexed: 12/20/2022] Open
Abstract
Background It has reported that long non-coding RNAs (lncRNAs) exerted regulatory functions by targeting specific genes through a competing endogenous RNA (ceRNA) pathway. LncRNA OIP5-AS1 has been identified as a tumor-enhancer in several tumor types. Nonetheless, its molecular mechanism in HCC remains to be masked. Aim of the study This study was aimed at exploring whether and how OIP5-AS1 exert functions in HCC. Methods qRT-PCR and western blot were employed for detecting gene expression. CCK-8, colony formation and EdU assays were implemented to evaluate the proliferative ability of HCC cells. Caspase-3 activity and flow cytometry analyses were implemented to determine cell apoptosis and cell cycle distribution. RNA pull down, ChIP, RIP and luciferase reporter assays explored the interplays between molecules. Results YY1 was upregulated in HCC cells, and silenced YY1 restrained HCC cell proliferation in vitro and hampered tumor growth in vivo. Later, we discovered that miR-300 could regulate WNT pathway via targeting YY1. Furthermore, OIP5-AS1 was identified as the sponge of miR-300 and promoted cell growth in HCC. Importantly, YY1 transcriptionally activate OIP5-AS1 in turn. Rescue experiments indicated that miR-300 inhibition or YY1 overexpression abrogated the inhibitive effect of OIP5-AS1 silencing on the malignant growth of HCC cells. Conclusions OIP5-AS1/miR-300/YY1 feedback loop facilitates cell growth in HCC by activating WNT pathway.
Collapse
Affiliation(s)
- Yu Wang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China.,Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Wuhan, 430030 Hubei China.,Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, 430030 Hubei China
| | - Lei Dou
- Department of Geratology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Yun Qin
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030 Hubei China
| | - Huiyuan Yang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| | - Peng Yan
- Department of Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 Hubei China
| |
Collapse
|
33
|
Liu HJ, Qin Y, Zhao ZH, Zhang Y, Yang JH, Zhai DH, Cui F, Luo C, Lu MX, Liu PP, Xu HW, Li K, Sun B, Chen S, Zhou HG, Yang C, Sun T. Lentinan-functionalized Selenium Nanoparticles target Tumor Cell Mitochondria via TLR4/TRAF3/MFN1 pathway. Theranostics 2020; 10:9083-9099. [PMID: 32802180 PMCID: PMC7415812 DOI: 10.7150/thno.46467] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 06/26/2020] [Indexed: 12/13/2022] Open
Abstract
Rationale: Malignant ascites caused by cancer cells results in poor prognosis and short average survival time. No effective treatment is currently available for malignant ascites. In this study, the effects of lentinan (LNT)-functionalized selenium nanoparticles (Selene) on malignant ascites were evaluated. Furthermore, the mechanism of Selene targeting mitochondria of tumor cells were also investigated. Methods: Selene were synthesized and characterized by TEM, AFM and particle size analysis. The OVCAR-3 and EAC cells induced ascites models were used to evaluate the effects of Selene on malignant ascites. Proteomic analysis, immunofluorescence, TEM and ICP-MS were used to determine the location of Selene in tumor cells. Mitochondrial membrane potential, ROS, ATP content, and caspase-1/3 activity were detected to evaluate the effect of Selene on mitochondrial function and cell apoptosis. Immunofluorescence, Co-IP, pull-down, duolink, Western blot, and FPLC were used to investigate the pathway of Selene targeting mitochondria. Results: Selene could effectively inhibit ascites induced by OVCAR-3 and EAC cells. Selene was mainly located in the mitochondria of tumor cells and induced apoptosis of tumor cells. The LNT in Selene was involved in caveolae-mediated endocytosis through the interaction between toll-like receptor-4 (TLR4) and caveolin 1 (CAV1). Furthermore, the Selene in the endocytic vesicles could enter the mitochondria via the mitochondrial membrane fusion pathway, which was mediated by TLR4/TNF receptor associated factor 3 (TRAF3)/mitofusin-1 (MFN1) protein complex. Conclusion: Selene is a candidate anticancer drug for the treatment of malignant ascites. And TLR4/TRAF3/MFN1 may be a specific nano-drug delivery pathway that could target the mitochondria.
Collapse
|
34
|
Wnt Signaling in Gynecologic Malignancies. Int J Mol Sci 2020; 21:ijms21124272. [PMID: 32560059 PMCID: PMC7348953 DOI: 10.3390/ijms21124272] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/12/2020] [Accepted: 06/13/2020] [Indexed: 12/24/2022] Open
Abstract
Gynecologic malignancies, including ovarian cancer, endometrial cancer, and cervical cancer, affect hundreds of thousands of women worldwide every year. Wnt signaling, specifically Wnt/β-catenin signaling, has been found to play an essential role in many oncogenic processes in gynecologic malignancies, including tumorigenesis, metastasis, recurrence, and chemotherapy resistance. As such, the Wnt/β-catenin signaling pathway has the potential to be a target for effective treatment, improving patient outcomes. In this review, we discuss the evidence supporting the importance of the Wnt signaling pathways in the development, progression, and treatment of gynecologic malignancies.
Collapse
|
35
|
Liu D, Song Z, Wang X, Ouyang L. Ubiquitin C-Terminal Hydrolase L5 (UCHL5) Accelerates the Growth of Endometrial Cancer via Activating the Wnt/β-Catenin Signaling Pathway. Front Oncol 2020; 10:865. [PMID: 32596150 PMCID: PMC7300206 DOI: 10.3389/fonc.2020.00865] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 05/01/2020] [Indexed: 01/22/2023] Open
Abstract
Endometrial cancer (EC) is the most prevalent gynecological malignancy with high mortality. Chemotherapy plays a pivotal role both in an adjuvant setting and in exclusive treatment. However, current pharmacotherapies are limited and not ideal for improving the overall survival of EC patients. Thus, identification of the underlying molecular mechanisms responsible for initiation and progression of EC is imperative for developing novel therapeutic strategies. Ubiquitin C-terminal hydrolase L5 (UCHL5) has been found to aggravate tumor growth and metastasis in several different types of tumor models such as esophageal squamous cell carcinoma, hepatocellular carcinoma, and epithelial ovarian cancer. However, whether UCHL5 influences the growth of EC has not been elucidated. To expose the role of UCHL5 on EC, bioinformatics analysis was conducted, and it hinted that UCHL5 was overexpressed in EC tissues and associated with lower overall survival. Consistently, the overexpression of UCHL5 in EC tissues and cell lines was further confirmed by western blot (WB) and polymerase chain reaction (PCR) compared with non-tumor control. Lentivirus vectors carrying UCHL5 shRNA or CD sequences were used to reduce or overexpress the UCHL5 gene, respectively. Cell proliferation and cycle were facilitated, and cell apoptosis was decreased when the UCHL5 gene was overexpressed in EC cell lines. These results were opposite in UCHL5 knockdown EC cells. Additionally, the expression of β-catenin is positively related to UCHL5 levels and the tumorigenic effects of UCHL5 overexpression were reversed by the Wnt/β-catenin pathway inhibitor XAV939. Thus, Wnt/β-catenin pathway activation may be a partial mechanism responsible for the promoting effects of UCHL5 on EC growth. In conclusion, UCHL5 accelerated the growth of EC via the Wnt/β-catenin pathway and was expected to be an attractive target for EC treatment.
Collapse
Affiliation(s)
- Da Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zixuan Song
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaoying Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ling Ouyang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|