1
|
Bagheri L, Javanbakht M, Malekian S, Ghahderijani BH, Taghipour S, Tanha FD, Ranjkesh M, Cegolon L, Zhao S. Antifibrotic therapeutic strategies in systemic sclerosis: Critical role of the Wnt/β-catenin and TGF-β signal transduction pathways as potential targets. Eur J Pharmacol 2025:177607. [PMID: 40209848 DOI: 10.1016/j.ejphar.2025.177607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
Systemic sclerosis (SSc) is a prototypic fibrosing disorder characterized by widespread fibrosis and immune dysregulation. Current evidence highlights the intricate cross-talk between the canonical Wnt/β-catenin signaling pathway and transforming growth factor-beta (TGF-β) signaling, both of which play fundamental roles in the pathogenesis of fibrosis. This review aims to elucidate the central role of the Wnt/β-catenin-TGF-β pathway and TGF-β signal transduction pathway in fibrotic diseases, focusing on SSc. We summarized evidence from cellular biology studies, animal model investigations, and clinical observations to provide a comprehensive view of the mechanisms by which these pathways cause pathological fibrosis. In addition, we explore the possibilities of antifibrotic therapeutic strategies against Wnt/β-catenin-TGF-β signaling to counteract fibrosis. We aim to delineate approaches towards effectively treating fibrosis in SSc by targeting these interconnected signaling pathways.
Collapse
Affiliation(s)
- Leyla Bagheri
- Department of Internal Medicine, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Javanbakht
- Nephrology and Urology Research Center, Clinical Science Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Sheida Malekian
- Department of Internal Medicine, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Sadra Taghipour
- Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Fatemeh Davari Tanha
- Department of Infertility, Yas Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Luca Cegolon
- Department of Medical, Surgical & Health Sciences, University of Trieste, 34128 Trieste, Italy; Public Health Department, University Health Agency Giuliano-Isontina (ASUGI), 34148 Trieste, Italy
| | - Shi Zhao
- School of Public Health, Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
2
|
Fan B, Pan Q, Yuan X, Du W, Yan Z. EIF2S2 as a prognostic marker and therapeutic target in glioblastoma: insights into its role and downstream mechanisms. Cancer Cell Int 2025; 25:126. [PMID: 40176031 PMCID: PMC11967041 DOI: 10.1186/s12935-025-03762-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 03/20/2025] [Indexed: 04/04/2025] Open
Abstract
Glioblastoma (GBM) the most common and most aggressive primary brain tumor has a five-year survival rate of less than 5%. The onset of GBM is very complicated and has always been the focus of researchers. This study analyzed data from 637 GBM and 20 normal tissues from The Cancer Genome Atlas (TCGA), and patients were categorized into high and low EIF2S2 expression groups. The Overall survival and disease-free survival of GBM patients in low expression of EIF2S2 group were significantly higher than those in high expression of EIF2S2 group (p < 0.001), and the expression level of EIF2S2 was significantly correlated with tumor grade (p < 0.001) and tumor recurrence (p < 0.001). The study designed three different short hairpin RNA (shRNA) sequence vectors, identifying shEIF2S2-1 as the most effective. This vector significantly reduced EIF2S2 expression, cell proliferation, and migration while increasing apoptosis in SHG-44 and U251 cells (p < 0.01). By detecting SHG-44 cells infected with shEIF2S2 vector and shCtrl with human whole gene expression chip, we identified WNT5A that is a downstream target gene of EIF2S2. Interfering with WNT5A and overexpressing EIF2S2 in SHG-44 and U251 cells revealed that EIF2S2 regulates WNT5A expression. This regulation led to an increased apoptosis rate (p < 0.05) and a significant reduction in cell migration (p < 0.05) in both the EIF2S2 overexpression and shWNT5A interference groups, confirming that WNT5A is a downstream regulatory target of EIF2S2. This study revealed the key role of EIF2S2 in GBM and its potential molecular mechanism.
Collapse
Affiliation(s)
- Bo Fan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, 050000, Shijiazhuang, Hebei, China
| | - Qing Pan
- Department of Hemodialysis, The Second Hospital of Hebei Medical University, No.215, Heping West Road, Xinhua District, 050000, Shijiazhuang, Hebei, China
| | - Xiaokai Yuan
- Department of Rehabilitation Medicine, The Second Hospital of Hebei Medical University, No.215, Heping West Road, Xinhua District, 050000, Shijiazhuang, Hebei, China
| | - Wei Du
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, 050000, Shijiazhuang, Hebei, China
| | - Zhongjie Yan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Xinhua District, 050000, Shijiazhuang, Hebei, China.
| |
Collapse
|
3
|
Wu S, Nasser B Singab A, Lin G, Wang Y, Zhu H, Yang G, Chen J, Li J, Li P, Zhao D, Tian J, Ye L. The regulatory role of integrin in gastric cancer tumor microenvironment and drug resistance. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 195:130-136. [PMID: 39798809 DOI: 10.1016/j.pbiomolbio.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/18/2024] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
Gastric cancer (GC) remains a significant global health burden due to its high aggressiveness, early metastasis, and poor prognosis. Despite advances in chemotherapy and targeted therapies, drug resistance remains a major obstacle to improving patient outcomes. Integrins, a family of transmembrane receptors, play a pivotal role in mediating tumor growth, invasion, and drug resistance by interacting with the tumor microenvironment (TME) and regulating signaling pathways such as Wnt/β-catenin, FAK, and MAPK. This review highlights the critical functions of various integrin subunits (e.g., α5, αv, β1, β3, β6) in promoting GC progression and their involvement in chemoresistance mechanisms. Additionally, integrins modulate immune cell infiltration and stromal cell interactions within the TME, further complicating GC treatment. Emerging evidence suggests that targeting integrins, either through inhibitors or integrin-specific therapeutic strategies, holds potential in overcoming drug resistance and improving clinical outcomes. This review underscores the need for further exploration of integrins as therapeutic targets in GC and suggests promising avenues for integrin-based therapies in personalized medicine.
Collapse
Affiliation(s)
- Songlin Wu
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Abdel Nasser B Singab
- Department of Pharmacognosy, Ain-Shams University, Cairo, Egypt; Centre of Drug Discovery Research and Development, Ain Shams University, Cairo, Egypt
| | - Guimei Lin
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; National Medical Products Administration Key Laboratory for Technology Research and Evaluation of Drug Products, Shandong University, Jinan, Shandong, China
| | - Yulu Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Huaibo Zhu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Guang Yang
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Jiaqi Chen
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Jiaxuan Li
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Peiyao Li
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Di Zhao
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Jing Tian
- Second Hospital of Shandong University, Jinan, Shandong, China
| | - Lan Ye
- Center for Cancer Prevention and Treatment, Second Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
4
|
Asghariazar V, Ojarood MV, Vatankhah MA, Panahizadeh R, Haris HM, Najafzadeh N, Khakbaz P, Soozangar N, Jeddi F. Metformin reverses 5-FU chemoresistance by downregulating DKK1, WNT5A, and ABCB1 expressions in gastric cancer: An experimental and bioinformatic study. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03879-5. [PMID: 39954066 DOI: 10.1007/s00210-025-03879-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
Gastric cancer (GC) is one of the most fatal types of solid neoplasms involving individuals globally due to its chemo-resistant and metastatic nature. 5-Fluorouracil (5-FU) resistance is a complex procedure concerning the prognosis of patients with GC treated with this agent. Metformin has recently been highlighted as a member of anti-diabetic agents for its potential anti-cancer influences. In this study, we investigated the chemo-sensitivity and cell death mechanisms by which metformin moderates its effects by targeting ABCB1 (MDR1), DKK1, WNT5A, and chemo-resistance proteins (P-gp and CD44) on 5-FU-resistant gastric cancer cells. MTT assay exhibited that the combined metformin treatment with 5-FU had a more cytotoxic effect than 5-FU alone. DAPI staining proved that metformin, 5-FU, and co-treatment of them exerted an apoptotic effect on GC cells. Immunocytochemistry illustrated that metformin and its combination with 5-fluorouracil reduced the protein expressions of CD44 and P-gp, compared to the control group. The outcomes of this research displayed that the co-treatment of metformin with 5-FU significantly diminished the expressions of ABCB1, WNT5A, and DKK1 in comparison to the control. The co-treatment of these agents also decreased the expression of WNT5A and ABCB1 compared to 5-FU alone group. Overall, this study's findings demonstrated that co-treating metformin with 5-FU could overcome chemoresistance in GC cells by reducing the expression of WNT5A, MDR1, P-gp, and CD44 levels.
Collapse
Affiliation(s)
- Vahid Asghariazar
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Deputy of Research & Technology, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Mohammad Amin Vatankhah
- Department of Radiology, Imam Reza Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
- Students Research Committee, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Panahizadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hamed Mohammadi Haris
- Division of Vascular and Endovascular Surgery, Department of Surgery, Shohada-Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nowruz Najafzadeh
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Parya Khakbaz
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Narges Soozangar
- Zoonoses Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Department of Genetics and Pathology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Farhad Jeddi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Department of Genetics and Pathology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
5
|
Pei S, Zhang D, Li Z, Liu J, Li Z, Chen J, Xie Z. The Role of the Fox Gene in Breast Cancer Progression. Int J Mol Sci 2025; 26:1415. [PMID: 40003882 PMCID: PMC11855465 DOI: 10.3390/ijms26041415] [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: 11/26/2024] [Revised: 01/25/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Forkhead box (FOX) genes are a family of transcription factors that participate in many biological activities, from early embryogenesis to the formation of organs, and from regulation of glucose metabolism to regulation of longevity. Given the extensive influence in the multicellular process, FOX family proteins are responsible for the progression of many types of cancers, especially lung cancer, breast cancer, prostate cancer, and other cancers. Breast cancer is the most common cancer among women, and 2.3 million women were diagnosed in 2020. So, various drugs targeting the FOX signaling pathway have been developed to inhibit breast cancer progression. While the role of the FOX family gene in cancer development has not received enough attention, discovering more potential drugs targeting the FOX signaling pathway is urgently demanded. Here, we review the main members in the FOX gene family and summarize their signaling pathway, including the regulation of the FOX genes and their effects on breast cancer progression. We hope this review will emphasize the understanding of the role of the FOX gene in breast cancer and inspire the discovery of effective anti-breast cancer medicines targeting the FOX gene in the future.
Collapse
Affiliation(s)
- Shaoxuan Pei
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Dechun Zhang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Zhuohan Li
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Jinkai Liu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Ziyi Li
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Jianrui Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Zhenzhen Xie
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
| |
Collapse
|
6
|
Nayak A, Streiff H, Gonzalez I, Adekoya OO, Silva I, Shenoy AK. Wnt Pathway-Targeted Therapy in Gastrointestinal Cancers: Integrating Benchside Insights with Bedside Applications. Cells 2025; 14:178. [PMID: 39936971 PMCID: PMC11816596 DOI: 10.3390/cells14030178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 02/13/2025] Open
Abstract
The Wnt signaling pathway is critical in the onset and progression of gastrointestinal (GI) cancers. Anomalies in this pathway, often stemming from mutations in critical components such as adenomatous polyposis coli (APC) or β-catenin, lead to uncontrolled cell proliferation and survival. In the case of colorectal cancer, dysregulation of the Wnt pathway drives tumor initiation and growth. Similarly, aberrant Wnt signaling contributes to tumor development, metastasis, and resistance to therapy in other GI cancers, such as gastric, pancreatic, and hepatocellular carcinomas. Targeting the Wnt pathway or its downstream effectors has emerged as a promising therapeutic strategy for combating these highly aggressive GI malignancies. Here, we review the dysregulation of the Wnt signaling pathway in the pathogenesis of GI cancers and further explore the therapeutic potential of targeting the various components of the Wnt pathway. Furthermore, we summarize and integrate the preclinical evidence supporting the therapeutic efficacy of potent Wnt pathway inhibitors with completed and ongoing clinical trials in GI cancers. Additionally, we discuss the challenges of Wnt pathway-targeted therapies in GI cancers to overcome these concerns for effective clinical translation.
Collapse
|
7
|
Liu B, Li X, Wang S, Jia H, Zhang X, Dong Q, Li J. Wnt5a promotes VM formation by modulating the stemness and EMT progression of prostate cancer cell. Transl Oncol 2025; 51:102155. [PMID: 39488007 PMCID: PMC11565535 DOI: 10.1016/j.tranon.2024.102155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/23/2024] [Accepted: 10/07/2024] [Indexed: 11/04/2024] Open
Abstract
The incidence of prostate cancer (PCa) is increasing annually, making it the leading cause of tumor-related mortality in males. The available treatment options for metastatic PCa are limited. Vasculogenic mimicry (VM), an emerging phenomenon involving aggressive tumor cells, has a significant impact on patient survival. Misregulation of Wnt5a expression is commonly observed during cancer progression. However, there is a lack of comprehensive studies investigating the effects of Wnt5a on tumor VM formation. In this study, we demonstrate that alterations in wnt5a expression, either through gain or loss, have a significant influence on the formation of VM in tumor cells mediated by cell stemness and EMT progression. Further research has demonstrated that Wnt5a regulates the formation of VM through the PI3K/JNK signaling pathway. These experimental findings offer a novel avenue for the clinical management of prostate cancer.
Collapse
Affiliation(s)
- Bide Liu
- Department of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China; Laboratory of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China
| | - Xun Li
- Department of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China; Laboratory of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China
| | - Shuheng Wang
- Department of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China; Laboratory of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China
| | - Hongliang Jia
- Department of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China; Laboratory of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China
| | - Xiaoan Zhang
- Department of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China; Laboratory of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China
| | - Qiang Dong
- Department of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China; Laboratory of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China
| | - Jiuzhi Li
- Department of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China; Laboratory of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830001, China.
| |
Collapse
|
8
|
Wei J, Wang X, Yu D, Tu Y, Yu Y. MicroRNA-mediated autophagy and drug resistance in cancer: mechanisms and therapeutic strategies. Discov Oncol 2024; 15:662. [PMID: 39549162 PMCID: PMC11569378 DOI: 10.1007/s12672-024-01525-9] [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: 07/29/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024] Open
Abstract
This paper provides an exhaustive overview of the intricate interplay between microRNAs (miRNAs) and autophagy in the context of human cancers, underscoring the pivotal role these non-coding RNAs play in modulating autophagic pathways and their implications for cancer development, progression, and resistance to therapy. MiRNAs, as critical regulators of gene expression post-transcription, influence various biological processes, including autophagy, a catabolic mechanism essential for cellular homeostasis, stress response, and survival. The review meticulously delineates the mechanisms through which miRNAs impact autophagy by targeting specific genes and signaling pathways, thereby affecting cancer cell proliferation, metastasis, and response to chemotherapy. It highlights several miRNAs with dual roles, acting either as oncogenes or tumor suppressors based on the cellular context and the specific autophagic pathways they regulate. The paper further explores the therapeutic potential of targeting miRNA-autophagy axis, offering insights into novel strategies for cancer treatment through modulation of this axis. Emphasizing the complexity of the miRNA-autophagy relationship, the review calls for more in-depth studies to unravel the nuanced regulatory networks between miRNAs and autophagy in cancer, which could pave the way for the development of innovative therapeutic interventions and diagnostic tools.
Collapse
Affiliation(s)
- Jinxing Wei
- Department of Neurosurgery, Brain Hospital Affiliated to Tongji University, No.2880, Qixin Road, Shanghai, China
| | - Xianghui Wang
- Department of Neurosurgery, Brain Hospital Affiliated to Tongji University, No.2880, Qixin Road, Shanghai, China
| | - Duo Yu
- Department of Biopharmaceutics School of Pharmacy, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yanyang Tu
- Research Center, The Huizhou Central People's Hospital, Guangdong Medical University, No. 41 Eling North Road, Huizhou, Guangdong, China.
| | - Yaoyu Yu
- Department of Neurosurgery, Brain Hospital Affiliated to Tongji University, No.2880, Qixin Road, Shanghai, China.
| |
Collapse
|
9
|
Abdallah MM, Yahia M, Tagelsir Ahmed Y, Alfaki M. WNT5A in Cancer: A Pan-Cancer Analysis Revealing Its Diagnostic and Prognostic Biomarker Potential. Cureus 2024; 16:e65190. [PMID: 39176352 PMCID: PMC11340256 DOI: 10.7759/cureus.65190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND AND OBJECTIVE The wingless-related integration site family (WNT) signaling pathway is critical for tumor progression and development. It is associated with various neoplasms produced by WNT pathway deregulation; WNT5A, a member of the WNT family, has been linked to carcinogenesis, exhibiting either oncogenic or tumor-suppressive effects. The study investigates how the gene affects certain types of cancer. The study aimed to evaluate the potential prognostic significance of WNT5A genes as diagnostic biomarkers for various types of cancer. METHODOLOGY We investigated WNT5A gene expression in a pan-cancer analysis using various bioinformatics databases, including GEPIA (Gene Expression Profiling Interactive Analysis), TIMER2 (Tumor Immune Estimation Resource, Version 2), the University of Alabama at Birmingham Cancer Data Analysis Portal UACLAN databases, the Kaplan-Meier (K-M) plotter, cBioPortal, and Gene Set Cancer Analysis (GSCA). We aimed to gain insight into the expression of WNT5A in various tumors and its relationship with immune infiltration, overall survival, and genetic changes. Public datasets validated WNT5A expression in lung squamous cell carcinoma (LUSC) and stomach adenocarcinoma (STAD) samples. RESULTS WNT5A pan-cancer analysis was highly expressed in two cancer types, including STAD and LUSC. Additionally, TIMER results showed a positive association of WNT5A with immune cell infiltration in LUSC and STAD. Survival analysis indicated that LUSC cancer exhibits better overall survival, while STAD has lower overall survival levels, which means a poor prognosis in the STAD cancer type. Furthermore, mutation analysis revealed that the WNT5A gene was mutated in 1.4% of cases, with most alterations being deletions followed by amplifications. CONCLUSIONS The WNT5A gene's high expression in many malignancies, including LUSC and STAD, suggests it could be used as a diagnostic biomarker. This study shows a relationship between WNT5A expression and immune cell abundance in LUSC and STAD. Our pan-cancer analysis of this gene is the first of its type, and it will inform future research, comprehensive investigation, and wet lab experiments.
Collapse
Affiliation(s)
| | - Mawada Yahia
- Medical Laboratory Science/Medical Microbiology, University of Khartoum, Khartoum, SDN
| | - Yousra Tagelsir Ahmed
- Microbiology, Faculty of Medical Laboratory Science, Alzaiem Alazhari University, Khartoum, SDN
| | | |
Collapse
|
10
|
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
|
11
|
Zou G, Huang Y, Zhang S, Ko KP, Kim B, Zhang J, Venkatesan V, Pizzi MP, Fan Y, Jun S, Niu N, Wang H, Song S, Ajani JA, Park JI. E-cadherin loss drives diffuse-type gastric tumorigenesis via EZH2-mediated reprogramming. J Exp Med 2024; 221:e20230561. [PMID: 38411616 PMCID: PMC10899090 DOI: 10.1084/jem.20230561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 09/27/2023] [Accepted: 01/29/2024] [Indexed: 02/28/2024] Open
Abstract
Diffuse-type gastric adenocarcinoma (DGAC) is a deadly cancer often diagnosed late and resistant to treatment. While hereditary DGAC is linked to CDH1 mutations, the role of CDH1/E-cadherin inactivation in sporadic DGAC tumorigenesis remains elusive. We discovered CDH1 inactivation in a subset of DGAC patient tumors. Analyzing single-cell transcriptomes in malignant ascites, we identified two DGAC subtypes: DGAC1 (CDH1 loss) and DGAC2 (lacking immune response). DGAC1 displayed distinct molecular signatures, activated DGAC-related pathways, and an abundance of exhausted T cells in ascites. Genetically engineered murine gastric organoids showed that Cdh1 knock-out (KO), KrasG12D, Trp53 KO (EKP) accelerates tumorigenesis with immune evasion compared with KrasG12D, Trp53 KO (KP). We also identified EZH2 as a key mediator promoting CDH1 loss-associated DGAC tumorigenesis. These findings highlight DGAC's molecular diversity and potential for personalized treatment in CDH1-inactivated patients.
Collapse
Affiliation(s)
- Gengyi Zou
- Division of Radiation Oncology, Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuanjian Huang
- Division of Radiation Oncology, Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shengzhe Zhang
- Division of Radiation Oncology, Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kyung-Pil Ko
- Division of Radiation Oncology, Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bongjun Kim
- Division of Radiation Oncology, Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jie Zhang
- Division of Radiation Oncology, Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vishwa Venkatesan
- Division of Radiation Oncology, Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Melissa P. Pizzi
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yibo Fan
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sohee Jun
- Division of Radiation Oncology, Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Na Niu
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Huamin Wang
- Division of Pathology/Lab Medicine, Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shumei Song
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jaffer A. Ajani
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jae-Il Park
- Division of Radiation Oncology, Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
12
|
Sasahara M, Kanda M, Tanaka C, Shimizu D, Umeda S, Takami H, Inokawa Y, Hattori N, Hayashi M, Nakayama G, Kodera Y. Therapeutic antibody targeting natriuretic peptide receptor 1 inhibits gastric cancer growth via BCL-2-mediated intrinsic apoptosis. Int J Cancer 2024; 154:1272-1284. [PMID: 38151776 DOI: 10.1002/ijc.34831] [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: 08/12/2023] [Revised: 11/13/2023] [Accepted: 12/06/2023] [Indexed: 12/29/2023]
Abstract
Despite recent advances in the development of therapeutic antibodies, the prognosis of unresectable or metastatic gastric cancer (GC) remains poor. Here, we searched for genes involved in the malignant phenotype of GC and investigated the potential of one candidate gene to serve as a novel therapeutic target. Analysis of transcriptome datasets of GC identified natriuretic peptide receptor 1 (NPR1), a plasma membrane protein, as a potential target. We employed a panel of human GC cell lines and gene-specific small interfering RNA-mediated NPR1 silencing to investigate the roles of NPR1 in malignancy-associated functions and intracellular signaling pathways. We generated an anti-NPR1 polyclonal antibody and examined its efficacy in a mouse xenograft model of GC peritoneal dissemination. Associations between NPR1 expression in GC tissue and clinicopathological factors were also evaluated. NPR1 mRNA was significantly upregulated in several GC cell lines compared with normal epithelial cells. NPR1 silencing attenuated GC cell proliferation, invasion, and migration, and additionally induced the intrinsic apoptosis pathway associated with mitochondrial dysfunction and caspase activation via downregulation of BCL-2. Administration of anti-NPR1 antibody significantly reduced the number and volume of GC peritoneal tumors in xenografted mice. High expression of NPR1 mRNA in clinical GC specimens was associated with a significantly higher rate of postoperative recurrence and poorer prognosis. NPR1 regulates the intrinsic apoptosis pathway and plays an important role in promoting the GC malignant phenotype. Inhibition of NPR1 with antibodies may have potential as a novel therapeutic modality for unresectable or metastatic GC.
Collapse
Affiliation(s)
- Masahiro Sasahara
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mitsuro Kanda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Chie Tanaka
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Dai Shimizu
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinichi Umeda
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hideki Takami
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshikuni Inokawa
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norifumi Hattori
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masamichi Hayashi
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Goro Nakayama
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
13
|
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
|
14
|
Huang Y, Xue Q, Chang J, Wang X, Miao C. Wnt5a: A promising therapeutic target for inflammation, especially rheumatoid arthritis. Cytokine 2023; 172:156381. [PMID: 37806072 DOI: 10.1016/j.cyto.2023.156381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/05/2023] [Accepted: 09/22/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Wnt5a is a member of the Wnt protein family, which acts on classical or multiple non-classical Wnt signaling pathways by binding to different receptors. The expression regulation and signal transduction of Wnt5a is closely related to the inflammatory response. Abnormal activation of Wnt5a signaling is an important part of inflammation and rheumatoid arthritis (RA). OBJECTIVES This paper mainly focuses on Wnt5a protein and its mediated signaling pathway, summarizes the latest research progress of Wnt5a in the pathological process of inflammation and RA, and looks forward to the main directions of Wnt5a in RA research, aiming to provide a theoretical basis for the prevention and treatment of RA diseases by targeting Wnt5a. RESULTS Wnt5a is highly expressed in activated blood vessels, histocytes and synoviocytes in inflammatory diseases such as sepsis, sepsis, atherosclerosis and rheumatoid arthritis. It mediates the production of pro-inflammatory cytokines and chemokines, regulates the migration and recruitment of various immune effector cells, and thus participates in the inflammatory response. Wnt5a plays a pathological role in synovial inflammation and bone destruction of RA, and may be an important clinical therapeutic target for RA. CONCLUSION Wnt5a is involved in the pathological process of inflammation and interacts with inflammatory factors. Wnt5a may be a new target for regulating the progression of RA disease and intervening therapy because of its multi-modal effects on the etiology of RA, especially as a regulator of osteoclast activity and inflammation.
Collapse
Affiliation(s)
- Yurong Huang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Qiuyun Xue
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Jun Chang
- Department of Orthopaedics, the First Affiliated Hospital, Anhui Medical University, Hefei 230032, China; Anhui Public Health Clinical Center, Hefei, China.
| | - Xiao Wang
- Department of Clinical Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China.
| | - Chenggui Miao
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.
| |
Collapse
|
15
|
Krzysiek-Maczka G, Brzozowski T, Ptak-Belowska A. Helicobacter pylori-activated fibroblasts as a silent partner in gastric cancer development. Cancer Metastasis Rev 2023; 42:1219-1256. [PMID: 37460910 PMCID: PMC10713772 DOI: 10.1007/s10555-023-10122-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/20/2023] [Indexed: 12/18/2023]
Abstract
The discovery of Helicobacter pylori (Hp) infection of gastric mucosa leading to active chronic gastritis, gastroduodenal ulcers, and MALT lymphoma laid the groundwork for understanding of the general relationship between chronic infection, inflammation, and cancer. Nevertheless, this sequence of events is still far from full understanding with new players and mediators being constantly identified. Originally, the Hp virulence factors affecting mainly gastric epithelium were proposed to contribute considerably to gastric inflammation, ulceration, and cancer. Furthermore, it has been shown that Hp possesses the ability to penetrate the mucus layer and directly interact with stroma components including fibroblasts and myofibroblasts. These cells, which are the source of biophysical and biochemical signals providing the proper balance between cell proliferation and differentiation within gastric epithelial stem cell compartment, when exposed to Hp, can convert into cancer-associated fibroblast (CAF) phenotype. The crosstalk between fibroblasts and myofibroblasts with gastric epithelial cells including stem/progenitor cell niche involves several pathways mediated by non-coding RNAs, Wnt, BMP, TGF-β, and Notch signaling ligands. The current review concentrates on the consequences of Hp-induced increase in gastric fibroblast and myofibroblast number, and their activation towards CAFs with the emphasis to the altered communication between mesenchymal and epithelial cell compartment, which may lead to inflammation, epithelial stem cell overproliferation, disturbed differentiation, and gradual gastric cancer development. Thus, Hp-activated fibroblasts may constitute the target for anti-cancer treatment and, importantly, for the pharmacotherapies diminishing their activation particularly at the early stages of Hp infection.
Collapse
Affiliation(s)
- Gracjana Krzysiek-Maczka
- Department of Physiology, the Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531, Kraków, Poland.
| | - Tomasz Brzozowski
- Department of Physiology, the Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531, Kraków, Poland.
| | - Agata Ptak-Belowska
- Department of Physiology, the Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegorzecka Street, 31-531, Kraków, Poland
| |
Collapse
|
16
|
Hirata Y, Noorani A, Song S, Wang L, Ajani JA. Early stage gastric adenocarcinoma: clinical and molecular landscapes. Nat Rev Clin Oncol 2023; 20:453-469. [PMID: 37264184 DOI: 10.1038/s41571-023-00767-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2023] [Indexed: 06/03/2023]
Abstract
Gastric adenocarcinoma, even when diagnosed at an early (localized) disease stage, poses a major health-care burden with cure rates that remain unsatisfactorily low, particularly in Western countries. This lack of progress reflects, among other aspects, the impracticality of early diagnosis, considerable variations in therapeutic approaches that is partly based on regional preferences, and the ingrained heterogeneity of gastric adenocarcinoma cells and their associated tumour microenvironment (TME). Clinical trials have long applied empirical interventions with the assumption that all early stage gastric adenocarcinomas are alike. Despite certain successes, the shortcomings of these approaches can potentially be overcome by targeting the specific molecular subsets of gastric adenocarcinomas identified by genomic and/or multi-omics analyses, including microsatellite instability-high, Epstein-Barr virus-induced, DNA damage repair-deficient, HER2-positive and PD-L1-high subtypes. Future approaches, including the availability of sophisticated vaccines, novel antibody technologies, agents targeting TME components (including fibroblasts, macrophages, cytokines or chemokines, and T cells) and novel immune checkpoint inhibitors, supported by improved tissue-based and blood-based diagnostic assays, seem promising. In this Review, we highlight current knowledge of the molecular and cellular biology of gastric adenocarcinomas, summarize the current approaches to clinical management of the disease, and consider the role of novel management and/or treatment strategies.
Collapse
Affiliation(s)
- Yuki Hirata
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ayesha Noorani
- Cancer Ageing and Somatic Mutation Group, Wellcome Sanger Institute, Hinxton, UK
- Cambridge Oesophago-gastric Centre, Addenbrooke's Hospital, Cambridge, UK
| | - Shumei Song
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
17
|
Rojas A, Lindner C, Schneider I, González I, Morales MA. Contributions of the receptor for advanced glycation end products axis activation in gastric cancer. World J Gastroenterol 2023; 29:997-1010. [PMID: 36844144 PMCID: PMC9950863 DOI: 10.3748/wjg.v29.i6.997] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/26/2022] [Accepted: 01/11/2023] [Indexed: 02/10/2023] Open
Abstract
Compelling shreds of evidence derived from both clinical and experimental research have demonstrated the crucial contribution of receptor for advanced glycation end products (RAGE) axis activation in the development of neoplasms, including gastric cancer (GC). This new actor in tumor biology plays an important role in the onset of a crucial and long-lasting inflammatory milieu, not only by supporting phenotypic changes favoring growth and dissemination of tumor cells, but also by functioning as a pattern-recognition receptor in the inflammatory response to Helicobacter pylori infection. In the present review, we aim to highlight how the overexpression and activation of the RAGE axis contributes to the proliferation and survival of GC cells as and their acquisition of more invasive phenotypes that promote dissemination and metastasis. Finally, the contribution of some single nucleotide polymorphisms in the RAGE gene as susceptibility or poor prognosis factors is also discussed.
Collapse
Affiliation(s)
- Armando Rojas
- Biomedical Research Laboratories, Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
| | - Cristian Lindner
- Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
| | - Iván Schneider
- Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
| | - Ileana González
- Biomedical Research Laboratories, Medicine Faculty, Catholic University of Maule, Talca 34600000, Chile
| | - Miguel Angel Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago 8320000, Chile
| |
Collapse
|
18
|
El-Mahdy HA, Elsakka EGE, El-Husseiny AA, Ismail A, Yehia AM, Abdelmaksoud NM, Elshimy RAA, Noshy M, Doghish AS. miRNAs role in bladder cancer pathogenesis and targeted therapy: Signaling pathways interplay - A review. Pathol Res Pract 2023; 242:154316. [PMID: 36682282 DOI: 10.1016/j.prp.2023.154316] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023]
Abstract
Bladder cancer (BC) is the 11th most popular cancer in females and 4th in males. A lot of efforts have been exerted to improve BC patients' care. Besides, new approaches have been developed to enhance the efficiency of BC diagnosis, prognosis, therapeutics, and monitoring. MicroRNAs (miRNAs, miRs) are small chain nucleic acids that can regulate wide networks of cellular events. They can inhibit or degrade their target protein-encoding genes. The miRNAs are either downregulated or upregulated in BC due to epigenetic alterations or biogenesis machinery abnormalities. In BC, dysregulation of miRNAs is associated with cell cycle arrest, apoptosis, proliferation, metastasis, treatment resistance, and other activities. A variety of miRNAs have been related to tumor kind, stage, or patient survival. Besides, although new approaches for using miRNAs in the diagnosis, prognosis, and treatment of BC have been developed, it still needs further investigations. In the next words, we illustrate the recent advances in the role of miRNAs in BC aspects. They include the role of miRNAs in BC pathogenesis and therapy. Besides, the clinical applications of miRNAs in BC diagnosis, prognosis, and treatment are also discussed.
Collapse
Affiliation(s)
- Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| | - Elsayed G E Elsakka
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Ahmed A El-Husseiny
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt; Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City 11829, Cairo, Egypt
| | - Ahmed Ismail
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Amr Mohamed Yehia
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Nourhan M Abdelmaksoud
- Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Reham A A Elshimy
- Clinical & Chemical Pathology Department, National Cancer Institute, Cairo University, 11796 Cairo, Egypt
| | - Mina Noshy
- Clinical Pharmacy Department, Faculty of Pharmacy, King Salman International University (KSIU), SouthSinai, Ras Sudr 46612, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| |
Collapse
|
19
|
Chen Q, Zheng X, Li Y, Ma B, Nie X, Li M, Liu Y, Xu J, Yang Y. Wnt5a regulates autophagy in Bacille Calmette-Guérin (BCG)-Infected pulmonary epithelial cells. Microb Pathog 2022; 173:105826. [PMID: 36243383 DOI: 10.1016/j.micpath.2022.105826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 09/10/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
Abstract
Autophagy functions as a critical process that can suppress the proliferation of Mycobacterium tuberculosis (Mtb) within infected host cells. Wnt5a is a secreted protein that plays a range of physiological functions, activating several signaling pathways and thereby controlling cellular responses to particular stimuli. The importance of Wnt5a as a regulator of protection against Mtb infection, however, has yet to be fully characterized. Here, changes in murine pulmonary epithelial-like TC-1 cell morphology, autophagy, the Wnt/Ca2+ signaling pathway, and the mTOR autophagy pathway were analyzed following infection with the Mtb model pathogen Bacille Calmette-Guerin (BCG) in order to understand the regulatory role of Wnt5a in this context. These experiments revealed that Wnt5a was upregulated and autophagy was enhanced in TC-1 cells infected with BCG, and Wnt5a overexpression was found to drive BCG-induced autophagy in these cells upon infection, whereas the inhibition or knockdown of Wnt5a yielded the opposite effect. At the mechanistic level, Wnt5a was found to mediate non-canonical Wnt/Ca2+ signaling and thereby inhibit mTOR-dependent pathway activation, promoting autophagic induction within BCG-infected TC-1 cells. These data offer new insight regarding how Wnt5a influences Mtb-induced autophagy within pulmonary epithelial cells, providing a foundation for further research exploring the immunological control of this infection through the modulation of autophagy.
Collapse
Affiliation(s)
- Qi Chen
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, Ningxia, 750021, China; School of Life Sciences, Ningxia University, Yinchuan, Ningxia, 750021, China
| | - Xuedi Zheng
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, Ningxia, 750021, China; School of Life Sciences, Ningxia University, Yinchuan, Ningxia, 750021, China
| | - Yong Li
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, Ningxia, 750021, China; School of Life Sciences, Ningxia University, Yinchuan, Ningxia, 750021, China
| | - Boli Ma
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, Ningxia, 750021, China; School of Life Sciences, Ningxia University, Yinchuan, Ningxia, 750021, China
| | - Xueyi Nie
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, Ningxia, 750021, China; School of Life Sciences, Ningxia University, Yinchuan, Ningxia, 750021, China
| | - Mengyuan Li
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, Ningxia, 750021, China; School of Life Sciences, Ningxia University, Yinchuan, Ningxia, 750021, China; Key Laboratory of Hui Ethnic Medicine Modernization Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China
| | - Yueyang Liu
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, Ningxia, 750021, China; School of Life Sciences, Ningxia University, Yinchuan, Ningxia, 750021, China
| | - Jinrui Xu
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, Ningxia, 750021, China; School of Life Sciences, Ningxia University, Yinchuan, Ningxia, 750021, China.
| | - Yi Yang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, Ningxia, 750021, China; School of Life Sciences, Ningxia University, Yinchuan, Ningxia, 750021, China.
| |
Collapse
|
20
|
Luo R. CircRNA circ-MYBL2 absorbs precursor miR-92b in the nucleus to suppress its role in enhancing gastric cancer cell proliferation. Am J Med Sci 2022; 364:454-460. [PMID: 35644224 DOI: 10.1016/j.amjms.2022.05.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 03/05/2022] [Accepted: 05/09/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND MM-associated circular RNA (Circ-MYBL2), a circular RNA (circRNA), participates in cancer biology. However, its role in gastric cancer (GC) is unclear. In this study, circ-MYBL2 was predicted to interact with precursor microRNA-92b (miR-92b). We then analyzed the role of circ-MYBL2 in GC and explored its crosstalk with miR-92b. METHODS In this research Circ-MYBL2 and miR-92b (mature and precursor) accumulation was determined using reverse transcription polymerase chain reaction (RT-qPCR). The involvement of circ-MYBL2 in the maturation of miR-92b was analyzed using overexpression assays. The subcellular location of circ-MYBL2 was determined using nuclear fractionation assay. The binding of precursor miR-92b to circ-MYBL2 was analyzed through RNA-RNA pulldown assay. The role of circ-MYBL2 and miR-92b in GC cell proliferation was studied with BrdU assay. RESULTS We found that GC tissues exhibited increased mature miR-92b levels but decreased precursor miR-92b and circ-MYBL2 levels. Circ-MYBL2 was detected in both the nucleus and cytoplasm in GC cells, and it directly interacted with precursor miR-92b. Moreover, circ-MYBL2 overexpression increased precursor miR-92b expression and decreased mature miR-92b level. Furthermore, miR-92b (mature) increased GC cell proliferation, and circ-MYBL2 decreased GC cell proliferation and suppressed the effect of miR-92b on GC cell proliferation. CONCLUSIONS Circ-MYBL2 may absorb precursor miR-92b in the nucleus to suppress its role in promoting gastric cancer cell proliferation.
Collapse
Affiliation(s)
- Ruijie Luo
- Department of Critical Care Medicine, Chengdu Wenjiang District People's Hospital, Chengdu City, Sichuan, China.
| |
Collapse
|
21
|
Behera JK, Bhattacharya M, Mishra P, Mishra A, Dash AA, Kar NB, Behera B, Patra BC. Regulatory role of miRNAs in Wnt signaling pathway linked with cardiovascular diseases. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 3:100133. [PMID: 36568258 PMCID: PMC9780067 DOI: 10.1016/j.crphar.2022.100133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/15/2022] [Accepted: 09/30/2022] [Indexed: 11/06/2022] Open
Abstract
MicroRNAs (miRNAs) are discovered in science about 23 years ago. These are short, a series of non-coding, single-stranded and evolutionary conserved RNA molecules found in eukaryotic cells. It involved post-transcriptional fine-tune protein expression and repressing the target of mRNA in different biological processes. These miRNAs binds with the 3'-UTR region of specific mRNAs to phosphorylate the mRNA degradation and inhibit the translation process in various tissues. Therefore, aberrant expression in miRNAs induces numerous cardiovascular diseases and developmental defects. Subsequently, the miRNAs and Wnt singling pathway are regulating a cellular process in cardiac development and regeneration, maintain the homeostasis and associated heart diseases. In Wnt signaling pathway majority of the signaling components are expressed and regulated by miRNAs, whereas the inhibition or dysfunction of the Wnt signaling pathway induces cardiovascular diseases. Moreover, inadequate studies about the important role of miRNAs in heart development and diseases through Wnt signaling pathway has been exist still now. For this reason in present review we summarize and update the involvement of miRNAs and the role of Wnt signaling in cardiovascular diseases. We have discussed the mechanism of miRNA functions which regulates the Wnt components in cellular signaling pathway. The fundamental understanding of Wnt signaling regulation and mechanisms of miRNAs is quite essential for study of heart development and related diseases. This approach definitely enlighten the future research to provide a new strategy for formulation of novel therapeutic approaches against cardiovascular diseases.
Collapse
Affiliation(s)
- Jiban Kumar Behera
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore, 756089, Odisha, India
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore, 756089, Odisha, India
| | - Pabitra Mishra
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore, 756089, Odisha, India
| | - Akansha Mishra
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore, 756089, Odisha, India
| | - Adya Anindita Dash
- Department of Biosciences and Biotechnology, Fakir Mohan University, Vyasa Vihar, Balasore, 756089, Odisha, India
| | - Niladri Bhusan Kar
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore, 756089, Odisha, India
| | - Bhaskar Behera
- Department of Biosciences and Biotechnology, Fakir Mohan University, Vyasa Vihar, Balasore, 756089, Odisha, India
| | - Bidhan Chandra Patra
- Department of Zoology, Vidyasagar University, Midnapore, 721102, West Bengal, India
| |
Collapse
|
22
|
Islas JF, Quiroz-Reyes AG, Delgado-Gonzalez P, Franco-Villarreal H, Delgado-Gallegos JL, Garza-Treviño EN, Gonzalez-Villarreal CA. Cancer Stem Cells in Tumor Microenvironment of Adenocarcinoma of the Stomach, Colon, and Rectum. Cancers (Basel) 2022; 14:3948. [PMID: 36010940 PMCID: PMC9405851 DOI: 10.3390/cancers14163948] [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: 06/30/2022] [Revised: 07/29/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
Gastrointestinal adenocarcinomas are one of the world's deadliest cancers. Cancer stem cells and the tissue microenvironment are highly regulated by cell and molecular mechanisms. Cancer stem cells are essential for maintenance and progression and are associated with resistance to conventional treatments. This article reviews the current knowledge of the role of the microenvironment during the primary establishment of gastrointestinal adenocarcinomas in the stomach, colon, and rectum and its relationship with cancer stem cells. We also describe novel developments in cancer therapeutics, such as targeted therapy, and discuss the advantages and disadvantages of different treatments for improving gastrointestinal cancer prognosis.
Collapse
Affiliation(s)
- Jose Francisco Islas
- Biochemistry and Molecular Medicine Department, School of Medicine, Universidad Autonoma de Nuevo Leon, Monterrey 64460, Mexico
| | - Adriana G. Quiroz-Reyes
- Biochemistry and Molecular Medicine Department, School of Medicine, Universidad Autonoma de Nuevo Leon, Monterrey 64460, Mexico
| | - Paulina Delgado-Gonzalez
- Biochemistry and Molecular Medicine Department, School of Medicine, Universidad Autonoma de Nuevo Leon, Monterrey 64460, Mexico
| | | | - Juan Luis Delgado-Gallegos
- Biochemistry and Molecular Medicine Department, School of Medicine, Universidad Autonoma de Nuevo Leon, Monterrey 64460, Mexico
| | - Elsa N. Garza-Treviño
- Biochemistry and Molecular Medicine Department, School of Medicine, Universidad Autonoma de Nuevo Leon, Monterrey 64460, Mexico
| | | |
Collapse
|
23
|
dos Santos MGP, Gatti da Silva GH, Nagasse HY, Fuziwara CS, Kimura ET, Coltri PP. hnRNP A1 and hnRNP C associate with miR-17 and miR-18 in thyroid cancer cells. FEBS Open Bio 2022; 12:1253-1264. [PMID: 35417090 PMCID: PMC9157402 DOI: 10.1002/2211-5463.13409] [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: 10/08/2021] [Revised: 03/03/2022] [Accepted: 04/12/2022] [Indexed: 11/18/2022] Open
Abstract
Heterogeneous nuclear ribonucleoproteins (hnRNPs) are essential players in the regulation of gene expression. The majority of the twenty different hnRNP proteins act through the modulation of pre-mRNA splicing. Most have been shown to regulate the expression of critical genes for the progression of tumorigenic processes and were also observed to be overexpressed in several types of cancer. Moreover, these proteins were described as essential components for the maturation of some microRNAs (miRNAs). In the human genome, over 70% of miRNAs are transcribed from introns; therefore, we hypothesized that regulatory proteins involved with splicing could be important for their maturation. Increased expression of the miR-17-92 cluster has already been shown to be related to the development of many cancers, such as thyroid, lung, and lymphoma. In this article, we show that overexpression of hnRNP A1 and hnRNP C in BCPAP thyroid cancer cells directly affects the expression of miR-17-92 miRNAs. Both proteins associate with the 5'-end of this cluster, strongly precipitate miRNAs miR-17 and miR-18a and upregulate the expression of miR-92a. Upon overexpression of these hnRNPs, BCPAP cells also show increased proliferation, migration, and invasion rates, suggesting upregulation of these proteins and miRNAs is related to an enhanced tumorigenic phenotype.
Collapse
Affiliation(s)
- Maria Gabriela Pereira dos Santos
- Departamento de Biologia Celular e do DesenvolvimentoInstituto de Ciências BiomédicasUniversidade de São PauloBrazil
- Present address:
National Center for Tumor Diseases (NCT) DresdenFetscherstraße 74Dresden01307Germany
| | | | - Helder Yudi Nagasse
- Departamento de Biologia Celular e do DesenvolvimentoInstituto de Ciências BiomédicasUniversidade de São PauloBrazil
| | - Cesar Seigi Fuziwara
- Departamento de Biologia Celular e do DesenvolvimentoInstituto de Ciências BiomédicasUniversidade de São PauloBrazil
| | - Edna T. Kimura
- Departamento de Biologia Celular e do DesenvolvimentoInstituto de Ciências BiomédicasUniversidade de São PauloBrazil
| | - Patricia Pereira Coltri
- Departamento de Biologia Celular e do DesenvolvimentoInstituto de Ciências BiomédicasUniversidade de São PauloBrazil
| |
Collapse
|
24
|
Lu Z, Zhou Y, Jing Q. Wnt5a-mediated autophagy promotes radiation resistance of nasopharyngeal carcinoma. J Cancer 2022; 13:2388-2396. [PMID: 35517407 PMCID: PMC9066197 DOI: 10.7150/jca.71526] [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: 01/28/2022] [Accepted: 04/04/2022] [Indexed: 12/24/2022] Open
Abstract
Wnt signaling pathways and autophagy play an essential role in tumor progression. Canonical Wnt signaling pathways in radiation resistance have been studied in the past, but it remains unclear whether the noncanonical Wnt signaling pathways can affect tumor radiation resistance through protective autophagy. Nasopharyngeal carcinoma, a particular subtype of head and neck squamous cell carcinoma, relies on radiation therapy. In this study, we found that radioactive rays could significantly promote the expression of Wnt noncanonical signaling pathways ligands in nasopharyngeal carcinoma, among which Wnt5A was the most markedly altered. We have demonstrated that Wnt5a can reduce the radiation sensitivity of nasopharyngeal carcinoma in vitro and in vitro experiments. Meanwhile, we found much more greater autophagosomes in overexpressed-Wnt5A nasopharyngeal carcinoma cells by electron microscopy. Further mechanism exploration revealed that Beclin1 is the main target of Wnt5A, and knocking down Beclin1 can partially reduce Wnt5a-induced radiation resistance. By studying Wnt5A-mediated protective autophagy in promoting radiation resistance in nasopharyngeal carcinoma cells, we hope that the Wnt5A and Beclin1 can become effective targets for overcoming radiation resistance in the future.
Collapse
Affiliation(s)
- Zhaoyi Lu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Yandan Zhou
- Changsha Aier Eye Hospital, Aier Eye Hospital Group, Changsha, Hunan,410000, People's Republic of China
| | - Qiancheng Jing
- The Affiliated Changsha Central Hospital, Department of Otolaryngology Head and Neck Surgery, Hengyang Medical School, University of South China. Changsha, Hunan, 410001, People's Republic of China
| |
Collapse
|
25
|
Feng Y, Wang Y, Guo K, Feng J, Shao C, Pan M, Ding P, Liu H, Duan H, Lu D, Wang Z, Zhang Y, Zhang Y, Han J, Li X, Yan X. The value of WNT5A as prognostic and immunological biomarker in pan-cancer. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:466. [PMID: 35571400 PMCID: PMC9096401 DOI: 10.21037/atm-22-1317] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/13/2022] [Indexed: 11/25/2022]
Abstract
Background Finding new immune-related biomarkers is one of the promising research directions for tumor immunotherapy. The WNT5A gene could stimulate the WNT pathway and regulate the progression of various tumors. Recent studies have partially revealed the relationship between WNT5A and tumor immunity, but the correlation and underlying mechanisms in pan-cancer remain obscure. Thus, we conducted this study aiming to characterize the prognostic value and immunological portrait of WNT5A in cancer. Methods The data obtained from The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), and Cancer Cell Line Encyclopedia (CCLE) databases was utilized to analyze WNT5A expression levels by Kruskal-Wallis test and correlation to prognosis by Cox regression test and Kaplan-Meier test, while the data was also used to study the association between WNT5A expression and immune microenvironment, immune neoantigens, immune checkpoints, tumor mutational burden (TMB), and microsatellite instability (MSI) in pan-cancer. Gene set enrichment analysis (GSEA) was used to clarify the relevant signaling pathways. The R package was used for data analysis and to create the plots. Results The pan-cancer analysis revealed that the expression level of WNT5A is generally elevated in most tumors (19/34, 55.88%), and high WNT5A expression was correlated with poor prognosis in esophageal carcinoma (ESCA, P<0.05), low-grade glioma (LGG, P<0.01), adrenocortical carcinoma (ACC, P<0.01), pancreatic adenocarcinoma (PAAD, P<0.01), and head and neck squamous cell carcinoma (HNSC, P<0.05). In addition, WNT5A expression was positively associated with immune infiltration, stromal score, and immune checkpoints in most cancers, and correlated to immune neoantigens, TMB, and MSI. Finally, GSEA indicated that WNT5A is implicated in the transforming growth factor β (TGFβ), Notch, and Hedgehog signaling pathways, which may be related to tumor immunity. Conclusions The expression of WNT5A is elevated in most tumors and associated with tumor prognosis. Furthermore, WNT5A is associated with tumor immunity and may be an immunological biomarker in cancer.
Collapse
Affiliation(s)
- Yingtong Feng
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China.,Department of Cardiothoracic Surgery, The 71st Group Army Hospital of PLA/The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yuanyong Wang
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Kai Guo
- Department of Thoracic Surgery, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Junjun Feng
- Department of Human Resource Management, The 71st Group Army Hospital of PLA/The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, China
| | - Changjian Shao
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Minghong Pan
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Peng Ding
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Honggang Liu
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Hongtao Duan
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Di Lu
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Zhaoyang Wang
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Yimeng Zhang
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Yujing Zhang
- Department of Cardiothoracic Surgery, The 71st Group Army Hospital of PLA/The Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jing Han
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Xiaofei Li
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi'an, China
| |
Collapse
|
26
|
Lei Y, Chen L, Liu J, Zhong Y, Deng L. The MicroRNA-Based Strategies to Combat Cancer Chemoresistance via Regulating Autophagy. Front Oncol 2022; 12:841625. [PMID: 35211417 PMCID: PMC8861360 DOI: 10.3389/fonc.2022.841625] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/13/2022] [Indexed: 12/12/2022] Open
Abstract
Chemoresistance frequently occurs in cancer treatment, which results in chemotherapy failure and is one of the most leading causes of cancer-related death worldwide. Understanding the mechanism of chemoresistance and exploring strategies to overcome chemoresistance have become an urgent need. Autophagy is a highly conserved self-degraded process in cells. The dual roles of autophagy (pro-death or pro-survival) have been implicated in cancers and chemotherapy. MicroRNA (miRNA) is a class of small non-coding molecules that regulate autophagy at the post-transcriptional level in cancer cells. The association between miRNAs and autophagy in cancer chemoresistance has been emphasized. In this review, we focus on the dual roles of miRNA-mediated autophagy in facilitating or combating chemoresistance, aiming to shed lights on the potential role of miRNAs as targets to overcome chemoresistance.
Collapse
Affiliation(s)
- Yuhe Lei
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Lei Chen
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Junshan Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yinqin Zhong
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Lijuan Deng
- Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
27
|
Kaneda T, Kurata T, Yoshida T, Kibata K, Yoshioka H, Yanagimoto H, Takeda K, Yoshida T, Tsuta K. Massive digital gene expression analysis reveals different predictive profiles for immune checkpoint inhibitor therapy between adenocarcinoma and squamous cell carcinoma of advanced lung cancer. BMC Cancer 2022; 22:154. [PMID: 35135489 PMCID: PMC8822674 DOI: 10.1186/s12885-022-09264-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 01/28/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors prolong the survival of non-small cell lung cancer (NSCLC) patients. Although it has been acknowledged that there is some correlation between the efficacy of anti-programmed cell death-1 (PD-1) antibody therapy and immunohistochemical analysis, this technique is not yet considered foolproof for predicting a favorable outcome of PD-1 antibody therapy. We aimed to predict the efficacy of nivolumab based on a comprehensive analysis of RNA expression at the gene level in advanced NSCLC. METHODS This was a retrospective study on patients with NSCLC who were administered nivolumab at the Kansai Medical University Hospital. To identify genes associated with response to anti-PD-1 antibodies, we grouped patients into responders (complete and partial response) and non-responders (stable and progressive disease) to nivolumab therapy. Significant genes were then identified for these groups using Welch's t-test. RESULTS Among 42 analyzed cases (20 adenocarcinomas and 22 squamous cell carcinomas), enhanced expression of MAGE-A4, BBC3, and OTOA genes was observed in responders with adenocarcinoma, and enhanced expression of DAB2, HLA-DPB,1 and CDH2 genes was observed in responders with squamous cell carcinoma. CONCLUSIONS This study predicted the efficacy of nivolumab based on a comprehensive analysis of mRNA expression at the gene level in advanced NSCLC. We also revealed different gene expression patterns as predictors of the effectiveness of anti PD-1 antibody therapy in adenocarcinoma and squamous cell carcinoma.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/immunology
- Adenocarcinoma/drug therapy
- Adenocarcinoma/immunology
- Adult
- Aged
- Aged, 80 and over
- Antigens, CD/immunology
- Antigens, Neoplasm/immunology
- Apoptosis Regulatory Proteins/immunology
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/immunology
- Cadherins/immunology
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/immunology
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/immunology
- Female
- GPI-Linked Proteins/immunology
- Gene Expression/drug effects
- Gene Expression/immunology
- HLA-DP beta-Chains/immunology
- Humans
- Immune Checkpoint Inhibitors/therapeutic use
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Male
- Middle Aged
- Neoplasm Proteins/immunology
- Nivolumab/therapeutic use
- Predictive Value of Tests
- Programmed Cell Death 1 Receptor/drug effects
- Programmed Cell Death 1 Receptor/immunology
- Proto-Oncogene Proteins/immunology
- RNA, Messenger/drug effects
- RNA, Messenger/immunology
- Retrospective Studies
- Treatment Outcome
Collapse
Affiliation(s)
- Toshihiko Kaneda
- Department of Thoracic Oncology, Kansai Medical University Hospital, 2-3-1, Shinmachi, Hirakata City, Osaka, 573-1191, Japan.
| | - Takayasu Kurata
- Department of Thoracic Oncology, Kansai Medical University Hospital, 2-3-1, Shinmachi, Hirakata City, Osaka, 573-1191, Japan
| | - Tomoko Yoshida
- Discovery Technology Research, Ono Pharmaceutical, Co., Ltd., Osaka, Japan
| | - Kayoko Kibata
- Department of Thoracic Oncology, Kansai Medical University Hospital, 2-3-1, Shinmachi, Hirakata City, Osaka, 573-1191, Japan
| | - Hiroshige Yoshioka
- Department of Thoracic Oncology, Kansai Medical University Hospital, 2-3-1, Shinmachi, Hirakata City, Osaka, 573-1191, Japan
| | - Hiroaki Yanagimoto
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
- Corporate Sponsored Research Programs for Cancer Immunogenomics, Kansai Medical University, Osaka, Japan
| | - Kazuhiko Takeda
- Research Center of Oncology, Ono Pharmaceutical, Co., Ltd., Osaka, Japan
| | - Takao Yoshida
- Research Center of Oncology, Ono Pharmaceutical, Co., Ltd., Osaka, Japan
| | - Koji Tsuta
- Corporate Sponsored Research Programs for Cancer Immunogenomics, Kansai Medical University, Osaka, Japan
- Department of Pathology, Kansai Medical University, Osaka, Japan
| |
Collapse
|
28
|
Sremac M, Paic F, Grubelic Ravic K, Serman L, Pavicic Dujmovic A, Brcic I, Krznaric Z, Nikuseva Martic T. Aberrant expression of SFRP1, SFRP3, DVL2 and DVL3 Wnt signaling pathway components in diffuse gastric carcinoma. Oncol Lett 2021; 22:822. [PMID: 34691249 PMCID: PMC8527567 DOI: 10.3892/ol.2021.13083] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/03/2021] [Indexed: 02/07/2023] Open
Abstract
Diffuse gastric carcinoma (DGC) is characterized by poorly cohesive cells, highly invasive growth patterns, poor prognosis and resistance to the majority of available systemic therapeutic strategies. It has been previously reported that the Wnt/β-catenin signaling pathway serves a prominent role in the tumorigenesis of gastric carcinoma. However, the mechanism underlying the dysregulation of this pathway in DGC has not been fully elucidated. Therefore, the present study aimed to investigate the expression profiles of Wnt antagonists, secreted frizzled-related protein 1 (SFRP1) and secreted frizzled-related protein 3 (SFRP3), and dishevelled protein family members, dishevelled segment polarity protein 2 (DVL2) and dishevelled segment polarity protein 3 (DVL3), in DGC tissues. The association between the expression levels of these factors and the clinicopathological parameters of the patients was determined. Protein and mRNA expression levels in 62 DGC tumor tissues and 62 normal gastric mucosal tissues obtained from patients with non-malignant disease were measured using immunohistochemical and reverse transcription-quantitative PCR (RT-qPCR) analysis. Significantly lower protein expression levels of SFRP1 (P<0.001) and SFRP3 (P<0.001), but significantly higher protein expression levels of DVL2 (P<0.001) and DVL3 (P<0.001) were observed in DGC tissues compared with in control tissues by immunohistochemistry. In addition, significantly lower expression levels of SFRP1 (P<0.05) and higher expression levels of DVL3 (P<0.05) were found in in DGC tissues compared with those in normal gastric mucosal tissues using RT-qPCR. According to correlation analysis between the SFRP1, SFRP3, DVL2 and DVL3 protein expression levels and the clinicopathological characteristics of patients with DGC, a statistically significant correlation was found between the SFRP3 volume density and T stage (r=0.304; P=0.017) and between the SFRP3 volume density and clinical stage (r=0.336; P=0.008). In conclusion, the findings of the present study suggested that the Wnt signaling pathway components SFRP1, SFRP3, DVL2 and DVL3 may be aberrantly expressed in DGC tissues, implicating their possible role in the development of this malignant disease. The present data also revealed a positive relationship between SFRP3 protein expression and the clinical and T stage of DGC.
Collapse
Affiliation(s)
- Maja Sremac
- Division of Gastroenterology and Hepatology, University Hospital Center, 10000 Zagreb, Croatia
| | - Frane Paic
- Laboratory for Epigenetics and Molecular Medicine, Department of Medical Biology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Katja Grubelic Ravic
- Division of Gastroenterology and Hepatology, University Hospital Center, 10000 Zagreb, Croatia
| | - Ljiljana Serman
- Department of Medical Biology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Centre of Excellence in Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Aja Pavicic Dujmovic
- Department of Radiology, General Hospital ‘Dr. Ivo Pedisic’, 44000 Sisak, Croatia
| | - Iva Brcic
- Diagnostic and Research Institute of Pathology, Medical University of Graz, A-8010 Graz, Austria
| | - Zeljko Krznaric
- Division of Gastroenterology and Hepatology, University Hospital Center, 10000 Zagreb, Croatia
| | - Tamara Nikuseva Martic
- Department of Medical Biology, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Centre of Excellence in Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
29
|
Astudillo P. An emergent Wnt5a/YAP/TAZ regulatory circuit and its possible role in cancer. Semin Cell Dev Biol 2021; 125:45-54. [PMID: 34764023 DOI: 10.1016/j.semcdb.2021.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 12/29/2022]
Abstract
Wnt5a is a ligand that plays several roles in development, homeostasis, and disease. A growing body of evidence indicates that Wnt5a is involved in cancer progression. Despite extensive research in this field, our knowledge about how Wnt5a is precisely involved in cancer is still incomplete. It is usually thought that certain combinations of Frizzled receptors and co-receptors might explain the observed effects of Wnt5a either as a tumor suppressor or by promoting migration and invasion. While accepting this 'receptor context' model, this review proposes that Wnt5a is integrated within a larger regulatory circuit involving β-catenin, YAP/TAZ, and LATS1/2. Remarkably, WNT5A and YAP1 are transcriptionally regulated by the Hippo and Wnt pathways, respectively, and might form a regulatory circuit acting through LATS kinases and secreted Wnt/β-catenin inhibitors, including Wnt5a itself. Therefore, understanding the precise role of Wnt5a and YAP in cancer requires a systems biology perspective.
Collapse
Affiliation(s)
- Pablo Astudillo
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile.
| |
Collapse
|
30
|
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
|
31
|
Khakbaz P, Panahizadeh R, Vatankhah MA, Najafzadeh N. Allicin Reduces 5-fluorouracil-resistance in Gastric Cancer Cells through Modulating MDR1, DKK1, and WNT5A Expression. Drug Res (Stuttg) 2021; 71:448-454. [PMID: 34261152 DOI: 10.1055/a-1525-1499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND & OBJECTIVE 5-fluorouracil (5-FU) is approved for the treatment of gastric carcinoma (GC), but chemo-resistance limits the application of it for GC. Thus, the combination of 5-FU with adjuvants such as allicin may overcome multidrug resistance (MDR). METHODS The anticancer effects of allicin, 5-FU, and allicin/5-FU on the 5-FU resistant MKN-45 cells were evaluated by MTT assay and DAPi staining. The expression of the P-glycoprotein (P-gp) and CD44 protein were determined using immunocytochemistry. We also quantified mRNA expression levels of WNT5A, Dickkopf-1 (DKK1), and MDR1 in the GC cells. RESULTS Here, we found that the combination of allicin with 5-FU significantly increased apoptosis compared to 5-FU alone (P<0.05). We showed that WNT5A, MDR1, and DKK1 mRNA expression levels were down-regulated in the allicin- and allicin/5-FU-treated cells. Indeed, the combination of allicin and 5-FU significantly decreased the expression of the P-gp and CD44 proteins (P<0.05). CONCLUSION Our findings indicate that the combination of allicin with 5-FU could reverse multidrug resistance in the GC cells by reducing the expression of WNT5A, DKK1, MDR1, P-gp, and CD44 levels.
Collapse
Affiliation(s)
- Parya Khakbaz
- Student Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Reza Panahizadeh
- Student Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Amin Vatankhah
- Student Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nowruz Najafzadeh
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
32
|
Nema R, Patel P, Kumar A. Prognostic Role of Receptor Tyrosine Kinase-Like Orphan Receptors in Intestinal-Type Gastric Cancer. Asian Pac J Cancer Prev 2021; 22:2125-2134. [PMID: 34319035 PMCID: PMC8607102 DOI: 10.31557/apjcp.2021.22.7.2125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/01/2021] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Gastric cancer (GC) is diagnosed at advanced stages and has high mortality rates. Surgical resection and adjuvant chemotherapy are the main therapeutic approaches for GC. Despite curative resection, recurrence and metastasis contribute to a high mortality rate in patients with GC. The receptor-tyrosine-kinase-like orphan receptors 1/2 (ROR1/2) are transmembrane proteins belonging to the receptor tyrosine kinase (RTK) family. ROR1 and ROR2 are known to overexpress in the tumor tissues from several types of cancer patients. However, the role of RORs in the prognosis has not been understood. METHODS This study aimed to determine the association of mRNA expression of ROR1, ROR2, and their signaling components WNT5A, NKX2-1, and FOXF1, with the survival outcome of GC patients. We performed Kaplan-Meir survival analysis on publicly available 'The Cancer Genome Atlas (TCGA)' data sets using 'Kaplan-Meir Plotter.' RESULTS High mRNA expression of ROR1, ROR2, NKX2-1, and FOXF1 was significantly correlated with worse overall survival (OS) of GC patients. Interestingly ROR1 and ROR showed a prognostic role in the intestinal subtype, but not in the diffuse subtype of GC. Furthermore, ROR1 was positively correlated with regulatory T cells and M2-type macrophages and negatively correlated with Th17 and natural killer T cells in the tumor stroma of patients with GC. CONCLUSION We conclude that the expression of ROR1, ROR2, and their associated genes correlate with worst prognosis of GC patients, particularly in the intestinal type. .
Collapse
Affiliation(s)
| | | | - Ashok Kumar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal, India.
| |
Collapse
|
33
|
Emerging role of lncRNAs in the regulation of Rho GTPase pathway. Biomed Pharmacother 2021; 140:111731. [PMID: 34015583 DOI: 10.1016/j.biopha.2021.111731] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 02/08/2023] Open
Abstract
The Ras homolog (Rho) family of small GTPases comprise several proteins with prominent roles in regulation of cell cycle transition, cell migration, and remodeling of actin cytoskeleton. Expression of these proteins is regulated by several factors among them are long non-coding RNAs (lncRNAs). The impact of lncRNAs on Rho GTPases signaling can be exerted through direct modulation of expression of these proteins or influencing expression of miRNAs that negatively regulate Rho GTPases. LINC00974/miR-122/RhoA, MALAT1/miR-429/RhoA, ZFAS1/miR-3924/RhoA/ROCK2, PCAT6/miR-326/RhoA/ROCK, SMILR/miR-141/RhoA/ROCK, DAPK1/miR-182/RhoA, GAS5/miR663a/RhoB, H19/miR-15b/CDC42/PAK1, TDRG1/miR-93/RhoC, TUG1/miR-498/CDC42, UCA1/miR-18a/Cdc42 and UCA1/miR-182/Cdc42 are examples of lncRNAs/miRNAs axes that regulate Rho GTPases. In the present manuscript, we describe the role of lncRNAs on Rho GTPases.
Collapse
|
34
|
Astudillo P. A Non-canonical Wnt Signature Correlates With Lower Survival in Gastric Cancer. Front Cell Dev Biol 2021; 9:633675. [PMID: 33869179 PMCID: PMC8047116 DOI: 10.3389/fcell.2021.633675] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 02/24/2021] [Indexed: 01/02/2023] Open
Abstract
Genetic evidence suggests a role for the Wnt/β-catenin pathway in gastric cancer. However, Wnt5a, regarded as a prototypical non-canonical Wnt ligand, has also been extensively associated with this disease. Therefore, the roles of the Wnt signaling pathway in gastric cancer initiation and progression, and particularly the precise mechanisms by which the non-canonical Wnt pathway might promote the development and progression of gastric cancer, are not entirely well understood. This article analyzes publicly available gene and protein expression data and reveals the existence of a WNT5A/FZD2/FZD7/ROR2 signature, which correlates with tumor-infiltrating and mesenchymal cell marker expression. High expression of FZD7 and ROR2 correlates with a shared gene and protein expression profile, which in turn correlates with poor prognosis. In summary, the findings presented in this article provide an updated view of the relative contributions of the Wnt/β-catenin and non-canonical Wnt pathways in gastric cancer.
Collapse
Affiliation(s)
- Pablo Astudillo
- Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| |
Collapse
|
35
|
Lymph node metastasis-derived gastric cancer cells educate bone marrow-derived mesenchymal stem cells via YAP signaling activation by exosomal Wnt5a. Oncogene 2021; 40:2296-2308. [PMID: 33654199 PMCID: PMC7994201 DOI: 10.1038/s41388-021-01722-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 02/14/2021] [Accepted: 02/18/2021] [Indexed: 12/14/2022]
Abstract
Lymph node metastasis (LNM), a common metastatic gastric-cancer (GC) route, is closely related to poor prognosis in GC patients. Bone marrow-derived mesenchymal stem cells (BM-MSCs) preferentially engraft at metastatic lesions. Whether BM-MSCs are specifically reprogrammed by LNM-derived GC cells (LNM-GCs) and incorporated into metastatic LN microenvironment to prompt GC malignant progression remains unknown. Herein, we found that LNM-GCs specifically educated BM-MSCs via secretory exosomes. Exosomal Wnt5a was identified as key protein mediating LNM-GCs education of BM-MSCs, which was verified by analysis of serum exosomes collected from GC patients with LNM. Wnt5a-enriched exosomes induced YAP dephosphorylation in BM-MSCs, whereas Wnt5a-deficient exosomes exerted the opposite effect. Inhibition of YAP signaling by verteporfin blocked LNM-GC exosome- and serum exosome-mediated reprogramming in BM-MSCs. Analysis of MSC-like cells obtained from metastatic LN tissues of GC patients (GLN-MSCs) confirmed that BM-MSCs incorporated into metastatic LN microenvironment, and that YAP activation participated in maintaining their tumor-promoting phenotype and function. Collectively, our results show that LNM-GCs specifically educated BM-MSCs via exosomal Wnt5a-elicited activation of YAP signaling. This study provides new insights into the mechanisms of LNM in GC and BM-MSC reprogramming, and will provide potential therapeutic targets and detection indicators for GC patients with LNM.
Collapse
|
36
|
Zhang Z, Liang L, Cao G. Critical role of miR-26a-5p/Wnt5a signaling in gambogic acid-induced inhibition of gastric cancer. J Biochem Mol Toxicol 2021; 35:e22721. [PMID: 33533530 DOI: 10.1002/jbt.22721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 10/06/2020] [Accepted: 11/26/2020] [Indexed: 12/24/2022]
Abstract
Gastric cancer (GC) represents the fifth most human malignant disease and the third-most common cause of cancer-related death. Gambogic acid (GA) is a natural compound with a polyprenylated xanthone structure and possesses remarkable antitumor activity in a variety of cancer cells. However, the mechanism underlying the inhibitory effect of GA in GC is far from being completely understood. The goal of the present study is to investigate whether potential microRNAs are involved in antitumor effect of GA toward GC and to elucidate the possible mechanisms. We identified that miR-26a-5p was significantly increased by GA in GC cell lines and xenograft tumor. Downregulation of miR-26a-5p not only prevented GA-induced inhibition on GC cell growth, but also suppressed GA-induced apoptosis of GC cells. Informatics assay predicted that Wnt5a was regulated by miR-26a-5p and GA-induced downregulation of Wnt5a was prevented by anti-miR-26a-5p. Reporter gene assay showed that miR-26a-5p could negatively regulate Wnt5a through direct binding with 3'-UTR messenger RNA of Wnt5a. Thus, upregulation of Wnt5a exhibited the same action tendency for GA-induced GC cell growth and apoptosis as observed by downregulation of miR-26a-5p. In conclusion, these results indicated that the inhibitory effect of GA on GC was mediated by the upregulation of miR-26a-5p and downregulation of Wnt5a. Our study provided new clues for the potential therapeutic effect of GA against GC and highlighted the importance of miR-26a-5p/Wnt5a pathway in the regulation of GC development.
Collapse
Affiliation(s)
- Zhixin Zhang
- Department of Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of Surgery, Shanxi Provincial Traditional Chinese Medicine Institute, Taiyuan, China
| | - Lili Liang
- Department of Dermatology, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Gang Cao
- Department of Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
37
|
Yadav V, Jobe N, Mehdawi L, Andersson T. Targeting Oncogenic WNT Signalling with WNT Signalling-Derived Peptides. Handb Exp Pharmacol 2021; 269:279-303. [PMID: 34455485 DOI: 10.1007/164_2021_528] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
WNT signalling is known to be a crucial regulator of embryonic development and tissue homeostasis. Aberrant expression of WNT signalling elements or their mutations has been implicated in carcinogenesis and/or the progression of several different cancer types. Investigations of how WNT signalling affects carcinogenesis and cancer progression have revealed that it has essential roles in the regulation of proliferation, apoptosis, and cancer stemness and in angiogenesis and metastasis. Consequently, WNT-targeted therapy has gained much attention and has resulted in the development of several small molecules, the majority of which act as inhibitors of different WNT signalling events. However, although numerous inhibitory WNT signalling drug candidates have been included in clinical trials, no significant breakthroughs have been made. This could possibly be due to problems with inefficient binding to the target, compensatory signalling mechanisms and toxicity towards normal cells. Therapeutic peptides targeting WNT signalling in cancer cells have been developed as an alternative approach, with the hope that they might overcome the limitations reported for small WNT inhibitory molecules. In this chapter, we describe recent developments made in the design and characterization of WNT signalling-derived peptides aiming at their use as alternative cancer therapeutics and/or combined adjuvant therapy to conventional therapies.
Collapse
Affiliation(s)
- Vikas Yadav
- Experimental Pathology, Department of Translational Medicine, Clinical Research Centre, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Njainday Jobe
- Experimental Pathology, Department of Translational Medicine, Clinical Research Centre, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Lubna Mehdawi
- Experimental Pathology, Department of Translational Medicine, Clinical Research Centre, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Tommy Andersson
- Experimental Pathology, Department of Translational Medicine, Clinical Research Centre, Lund University, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
38
|
Yin D, Lu X. Silencing of long non-coding RNA HCP5 inhibits proliferation, invasion, migration, and promotes apoptosis via regulation of miR-299-3p/SMAD5 axis in gastric cancer cells. Bioengineered 2020; 12:225-239. [PMID: 33371778 PMCID: PMC8806318 DOI: 10.1080/21655979.2020.1863619] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Gastric cancer (GC) is a common malignant gastrointestinal tumor with high mortality. Previous study has reported that the overexpression of lncRNA HCP5 was observed in gastric cancer tissues. The purpose of this study was to investigate the molecular mechanism underlying the effect of lncRNA HCP5 on the proliferative, migratory, and invasive abilities of GC cells. The relative mRNA expression of HCP5, miR-299-3p, and SMAD5 were determined by RT-qPCR. The expressions of proteins associated with apoptosis and invasion were detected by western blot. The interaction of HCP5 with miR-299-3p and SMAD5 with miR-299-3p was confirmed by luciferase reporter assay. The cellular behaviors of AGS cells were, respectively, detected by CCK-8 assays, colony formation assays, migration and invasion assays, and flow cytometry. In our study, lncRNA HCP5 was highly expressed in GC cell lines compared with normal gastric epithelial cell. LncRNA HCP5 silencing inhibited AGS cells proliferation, migration, and invasion, while promoted cell apoptosis. Moreover, miR-299-3p downregulation could abolish the effect of HCP5 knockdown on cellular behaviors of AGS cells. Interestingly, SMAD5 is identified as the downstream target of miR-299-3p, and its expression was inhibited by miR-299-3p. More importantly, SMAD5 silencing inhibited proliferation, migration, and invasion of GC cells, and promoted cell apoptosis. In a word, lncRNA HCP5 silencing inhibits GC cell proliferation, invasion, and migration while promoting its apoptosis via regulation of miR-299-3p/SMAD5 axis. Hence, lncRNA HCP5 could be a novel and promising target for GC treatment.
Collapse
Affiliation(s)
- Derong Yin
- Department of Gastroenterology, Hanzhong People's Hospital , Hanzhong, Shaanxi, PR China
| | - Xiaohong Lu
- Department of Gastroenterology, Hubei Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University , Wuhan, Hubei, PR China
| |
Collapse
|
39
|
Vinasco-Sandoval T, Moreira FC, F. Vidal A, Pinto P, Ribeiro-dos-Santos AM, Cruz RLS, Fonseca Cabral G, Anaissi AKM, Lopes KDP, Ribeiro-dos-Santos A, Demachki S, de Assumpção PP, Ribeiro-dos-Santos Â, Santos S. Global Analyses of Expressed Piwi-Interacting RNAs in Gastric Cancer. Int J Mol Sci 2020; 21:E7656. [PMID: 33081152 PMCID: PMC7593925 DOI: 10.3390/ijms21207656] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/07/2020] [Accepted: 08/18/2020] [Indexed: 12/15/2022] Open
Abstract
Gastric cancer (GC) represents a notable amount of morbidity and mortality worldwide. Understanding the molecular basis of CG will offer insight into its pathogenesis in an attempt to identify new molecular biomarkers to early diagnose this disease. Therefore, studies involving small non-coding RNAs have been widely explored. Among these, PIWI-interacting RNAs (piRNAs) are an emergent class that can play important roles in carcinogenesis. In this study, small-RNA sequencing was used to identify the global piRNAs expression profile (piRNome) of gastric cancer patients. We found 698 piRNAs in gastric tissues, 14 of which were differentially expressed (DE) between gastric cancer (GC), adjacent to gastric cancer (ADJ), and non-cancer tissues (NC). Moreover, three of these DE piRNAs (piR-48966*, piR-49145, piR-31335*) were differently expressed in both GC and ADJ samples in comparison to NC samples, indicating that the tumor-adjacent tissue was molecularly altered and should not be considered as a normal control. These three piRNAs are potential risk biomarkers for GC, especially piR-48966* and piR-31335*. Furthermore, an in-silico search for mRNAs targeted by the differentially expressed piRNAs revealed that these piRNAs may regulate genes that participate in cancer-related pathways, suggesting that these small non-coding RNAs may be directly and indirectly involved in gastric carcinogenesis.
Collapse
Affiliation(s)
- Tatiana Vinasco-Sandoval
- Graduate Program in Genetics and Molecular Biology, Laboratory of Human and Medical Genetics, Federal University of Pará, Belém, Pará 66075-110, Brazil; (T.V.-S.); (F.C.M.); (A.F.V.); (P.P.); (A.M.R.-d.-S.); (R.L.S.C.); (G.F.C.); (K.d.P.L.); (A.R.-d.-S.); (Â.R.-d.-S.)
| | - Fabiano Cordeiro Moreira
- Graduate Program in Genetics and Molecular Biology, Laboratory of Human and Medical Genetics, Federal University of Pará, Belém, Pará 66075-110, Brazil; (T.V.-S.); (F.C.M.); (A.F.V.); (P.P.); (A.M.R.-d.-S.); (R.L.S.C.); (G.F.C.); (K.d.P.L.); (A.R.-d.-S.); (Â.R.-d.-S.)
- Graduate Program in Oncology and Medical Sciences, Center of Oncology Research, Federal University of Pará, Belém, Pará 66063-023, Brazil; (A.K.M.A.); (S.D.); (P.P.d.A.)
| | - Amanda F. Vidal
- Graduate Program in Genetics and Molecular Biology, Laboratory of Human and Medical Genetics, Federal University of Pará, Belém, Pará 66075-110, Brazil; (T.V.-S.); (F.C.M.); (A.F.V.); (P.P.); (A.M.R.-d.-S.); (R.L.S.C.); (G.F.C.); (K.d.P.L.); (A.R.-d.-S.); (Â.R.-d.-S.)
| | - Pablo Pinto
- Graduate Program in Genetics and Molecular Biology, Laboratory of Human and Medical Genetics, Federal University of Pará, Belém, Pará 66075-110, Brazil; (T.V.-S.); (F.C.M.); (A.F.V.); (P.P.); (A.M.R.-d.-S.); (R.L.S.C.); (G.F.C.); (K.d.P.L.); (A.R.-d.-S.); (Â.R.-d.-S.)
- Graduate Program in Oncology and Medical Sciences, Center of Oncology Research, Federal University of Pará, Belém, Pará 66063-023, Brazil; (A.K.M.A.); (S.D.); (P.P.d.A.)
| | - André M. Ribeiro-dos-Santos
- Graduate Program in Genetics and Molecular Biology, Laboratory of Human and Medical Genetics, Federal University of Pará, Belém, Pará 66075-110, Brazil; (T.V.-S.); (F.C.M.); (A.F.V.); (P.P.); (A.M.R.-d.-S.); (R.L.S.C.); (G.F.C.); (K.d.P.L.); (A.R.-d.-S.); (Â.R.-d.-S.)
| | - Rebecca L. S. Cruz
- Graduate Program in Genetics and Molecular Biology, Laboratory of Human and Medical Genetics, Federal University of Pará, Belém, Pará 66075-110, Brazil; (T.V.-S.); (F.C.M.); (A.F.V.); (P.P.); (A.M.R.-d.-S.); (R.L.S.C.); (G.F.C.); (K.d.P.L.); (A.R.-d.-S.); (Â.R.-d.-S.)
| | - Gleyce Fonseca Cabral
- Graduate Program in Genetics and Molecular Biology, Laboratory of Human and Medical Genetics, Federal University of Pará, Belém, Pará 66075-110, Brazil; (T.V.-S.); (F.C.M.); (A.F.V.); (P.P.); (A.M.R.-d.-S.); (R.L.S.C.); (G.F.C.); (K.d.P.L.); (A.R.-d.-S.); (Â.R.-d.-S.)
| | - Ana K. M. Anaissi
- Graduate Program in Oncology and Medical Sciences, Center of Oncology Research, Federal University of Pará, Belém, Pará 66063-023, Brazil; (A.K.M.A.); (S.D.); (P.P.d.A.)
| | - Katia de Paiva Lopes
- Graduate Program in Genetics and Molecular Biology, Laboratory of Human and Medical Genetics, Federal University of Pará, Belém, Pará 66075-110, Brazil; (T.V.-S.); (F.C.M.); (A.F.V.); (P.P.); (A.M.R.-d.-S.); (R.L.S.C.); (G.F.C.); (K.d.P.L.); (A.R.-d.-S.); (Â.R.-d.-S.)
| | - Arthur Ribeiro-dos-Santos
- Graduate Program in Genetics and Molecular Biology, Laboratory of Human and Medical Genetics, Federal University of Pará, Belém, Pará 66075-110, Brazil; (T.V.-S.); (F.C.M.); (A.F.V.); (P.P.); (A.M.R.-d.-S.); (R.L.S.C.); (G.F.C.); (K.d.P.L.); (A.R.-d.-S.); (Â.R.-d.-S.)
| | - Samia Demachki
- Graduate Program in Oncology and Medical Sciences, Center of Oncology Research, Federal University of Pará, Belém, Pará 66063-023, Brazil; (A.K.M.A.); (S.D.); (P.P.d.A.)
| | - Paulo Pimentel de Assumpção
- Graduate Program in Oncology and Medical Sciences, Center of Oncology Research, Federal University of Pará, Belém, Pará 66063-023, Brazil; (A.K.M.A.); (S.D.); (P.P.d.A.)
| | - Ândrea Ribeiro-dos-Santos
- Graduate Program in Genetics and Molecular Biology, Laboratory of Human and Medical Genetics, Federal University of Pará, Belém, Pará 66075-110, Brazil; (T.V.-S.); (F.C.M.); (A.F.V.); (P.P.); (A.M.R.-d.-S.); (R.L.S.C.); (G.F.C.); (K.d.P.L.); (A.R.-d.-S.); (Â.R.-d.-S.)
- Graduate Program in Oncology and Medical Sciences, Center of Oncology Research, Federal University of Pará, Belém, Pará 66063-023, Brazil; (A.K.M.A.); (S.D.); (P.P.d.A.)
| | - Sidney Santos
- Graduate Program in Genetics and Molecular Biology, Laboratory of Human and Medical Genetics, Federal University of Pará, Belém, Pará 66075-110, Brazil; (T.V.-S.); (F.C.M.); (A.F.V.); (P.P.); (A.M.R.-d.-S.); (R.L.S.C.); (G.F.C.); (K.d.P.L.); (A.R.-d.-S.); (Â.R.-d.-S.)
- Graduate Program in Oncology and Medical Sciences, Center of Oncology Research, Federal University of Pará, Belém, Pará 66063-023, Brazil; (A.K.M.A.); (S.D.); (P.P.d.A.)
| |
Collapse
|
40
|
Lei Y, Chen L, Zhang G, Shan A, Ye C, Liang B, Sun J, Liao X, Zhu C, Chen Y, Wang J, Zhang E, Deng L. MicroRNAs target the Wnt/β‑catenin signaling pathway to regulate epithelial‑mesenchymal transition in cancer (Review). Oncol Rep 2020; 44:1299-1313. [PMID: 32700744 PMCID: PMC7448411 DOI: 10.3892/or.2020.7703] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/24/2020] [Indexed: 12/11/2022] Open
Abstract
Epithelial‑mesenchymal transition (EMT), during which cancer cells lose the epithelial phenotype and gain the mesenchymal phenotype, has been verified to result in tumor migration and invasion. Numerous studies have shown that dysregulation of the Wnt/β‑catenin signaling pathway gives rise to EMT, which is characterized by nuclear translocation of β‑catenin and E‑cadherin suppression. Wnt/β‑catenin signaling was confirmed to be affected by microRNAs (miRNAs), several of which are down‑ or upregulated in metastatic cancer cells, indicating their complex roles in Wnt/β‑catenin signaling. In this review, we demonstrated the targets of various miRNAs in altering Wnt/β‑catenin signaling to promote or inhibit EMT, which may elucidate the underlying mechanism of EMT regulation by miRNAs and provide evidence for potential therapeutic targets in the treatment of invasive tumors.
Collapse
Affiliation(s)
- Yuhe Lei
- Department of Pharmacy, Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518000, P.R. China
| | - Lei Chen
- Department of Pharmacy, Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518000, P.R. China
| | - Ge Zhang
- Department of Big Data Research of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong 510120, P.R. China
| | - Aiyun Shan
- Department of Pharmacy, Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518000, P.R. China
| | - Chunfeng Ye
- Department of Pediatrics, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Bin Liang
- Formula Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Jiayu Sun
- Department of Pharmacy, Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518000, P.R. China
| | - Xin Liao
- Department of Pharmacy, Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518000, P.R. China
| | - Changfeng Zhu
- Department of Pharmacy, Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518000, P.R. China
| | - Yueyue Chen
- Formula Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Jing Wang
- Formula Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Enxin Zhang
- Department of Oncology, Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518000, P.R. China
| | - Lijuan Deng
- Formula Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong 510632, P.R. China
| |
Collapse
|
41
|
Koushyar S, Powell AG, Vincan E, Phesse TJ. Targeting Wnt Signaling for the Treatment of Gastric Cancer. Int J Mol Sci 2020; 21:E3927. [PMID: 32486243 PMCID: PMC7311964 DOI: 10.3390/ijms21113927] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023] Open
Abstract
The Wnt signaling pathway is evolutionarily conserved, regulating both embryonic development and maintaining adult tissue homeostasis. Wnt signaling controls several fundamental cell functions, including proliferation, differentiation, migration, and stemness. It therefore plays an important role in the epithelial homeostasis and regeneration of the gastrointestinal tract. Often, both hypo- or hyper-activation of the pathway due to genetic, epigenetic, or receptor/ligand alterations are seen in many solid cancers, such as breast, colorectal, gastric, and prostate. Gastric cancer (GC) is the fourth commonest cause of cancer worldwide and is the second leading cause of cancer-related death annually. Although the number of new diagnoses has declined over recent decades, prognosis remains poor, with only 15% surviving to five years. Geographical differences in clinicopathological features are also apparent, with epidemiological and genetic studies revealing GC to be a highly heterogeneous disease with phenotypic diversity as a result of etiological factors. The molecular heterogeneity associated with GC dictates that a single 'one size fits all' approach to management is unlikely to be successful. Wnt pathway dysregulation has been observed in approximately 50% of GC tumors and may offer a novel therapeutic target for patients who would otherwise have a poor outcome. This mini review will highlight some recent discoveries involving Wnt signaling in GC.
Collapse
Affiliation(s)
- Sarah Koushyar
- European Cancer Stem Cell Research Institute, Cardiff University, Cardiff CF24 4HQ, UK; (S.K.); (A.G.P.)
| | - Arfon G. Powell
- European Cancer Stem Cell Research Institute, Cardiff University, Cardiff CF24 4HQ, UK; (S.K.); (A.G.P.)
- Division of Cancer & Genetics, Cardiff University, Cardiff CF14 4XW, UK
| | - Elizabeth Vincan
- Victorian Infectious Diseases Reference Laboratory, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia;
- School of Pharmacy and Biomedical Sciences, Curtin University, Perth WA 6102, Australia
| | - Toby J. Phesse
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne VIC 3000, Australia
| |
Collapse
|
42
|
Extracellular matrix stiffness and Wnt/β-catenin signaling in physiology and disease. Biochem Soc Trans 2020; 48:1187-1198. [DOI: 10.1042/bst20200026] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 12/11/2022]
Abstract
The Wnt/β-catenin signaling pathway plays fundamental roles during development, stem cell differentiation, and homeostasis, and its abnormal activation can lead to diseases. In recent years, it has become clear that this pathway integrates signals not only from Wnt ligands but also from other proteins and signaling routes. For instance, Wnt/β-catenin signaling involves YAP and TAZ, which are transcription factors with crucial roles in mechanotransduction. On the other hand, Wnt/β-catenin signaling is also modulated by integrins. Therefore, mechanical signals might similarly modulate the Wnt/β-catenin pathway. However, and despite the relevance that mechanosensitive Wnt/β-catenin signaling might have during physiology and diseases such as cancer, the role of mechanical cues on Wnt/β-catenin signaling has received less attention. This review aims to summarize recent evidence regarding the modulation of the Wnt/β-catenin signaling by a specific type of mechanical signal, the stiffness of the extracellular matrix. The review shows that mechanical stiffness can indeed modulate this pathway in several cell types, through differential expression of Wnt ligands, receptors and inhibitors, as well as by modulating β-catenin levels. However, the specific mechanisms are yet to be fully elucidated.
Collapse
|