1
|
Panahizadeh R, Panahi P, Asghariazar V, Makaremi S, Noorkhajavi G, Safarzadeh E. A literature review of recent advances in gastric cancer treatment: exploring the cross-talk between targeted therapies. Cancer Cell Int 2025; 25:23. [PMID: 39856676 PMCID: PMC11762578 DOI: 10.1186/s12935-025-03655-8] [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: 06/08/2024] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) ranks fourth in global mortality rates and fifth in prevalence, making it one of the most common cancers worldwide. Recent clinical studies have highlighted the potential of immunotherapies as a promising approach to treating GC. This study aims to shed light on the most impactful therapeutic strategies in the context of GC immunotherapy, highlighting both established and emerging approaches. MAIN BODY This review examines over 160 clinical studies conducted globally, focusing on the effectiveness of various immunotherapy modalities, including cancer vaccines, adoptive cell therapy, immune checkpoint inhibitors (ICIs), and monoclonal antibodies (mAbs). A comprehensive search of peer-reviewed literature was performed using databases such as Web of Science, PubMed, and Scopus. The selection criteria included peer-reviewed articles published primarily within the last 10 years, with a focus on studies that provided insights into targeted therapies and their mechanisms of action, clinical efficacy, and safety profiles. The findings indicate that these immunotherapy strategies can enhance treatment outcomes for GC, aligning with current treatment guidelines. ICIs like pembrolizumab and nivolumab have shown significant survival benefits in specific GC subgroups. Cancer vaccines and CAR-T cell therapies demonstrate potential, while mAbs targeting HER2 and VEGFR pathways enhance outcomes in combination regimens. We discuss the latest advancements and challenges in targeted therapy and immunotherapy for GC. Given the evolving nature of this field, this research emphasizes significant evidence-based therapies and those currently under evaluation rather than providing an exhaustive overview. Challenges include resistance mechanisms, immunosuppressive tumor environments, and inconsistent results from combination therapies. Biomarker-driven approaches and further research into emerging modalities like CAR-T cells and cancer vaccines are critical for optimizing treatments. CONCLUSIONS Immunotherapy is reshaping GC management by improving survival and quality of life. Ongoing research and clinical evaluations are crucial for refining personalized and effective therapies.
Collapse
Affiliation(s)
- Reza Panahizadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Padideh Panahi
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Asghariazar
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Shima Makaremi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ghasem Noorkhajavi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Department of Microbiology, Parasitology and Immunology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, 85991-56189, Iran.
| |
Collapse
|
2
|
Thilakasiri P, O'Keefe RN, To SQ, Chisanga D, Eissmann MF, Carli ALE, Duscio B, Baloyan D, Dmello RS, Williams D, Mariadason J, Poh AR, Pal B, Kile BT, Vissers JH, Harvey KF, Buchert M, Shi W, Ernst M, Chand AL. Mechanisms of cellular crosstalk in the gastric tumor microenvironment are mediated by YAP1 and STAT3. Life Sci Alliance 2024; 7:e202302411. [PMID: 37957015 PMCID: PMC10643184 DOI: 10.26508/lsa.202302411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/16/2023] Open
Abstract
Deregulation of the Hippo pathway is a driver for cancer progression and treatment resistance. In the context of gastric cancer, YAP1 is a biomarker for poor patient prognosis. Although genomic tumor profiling provides information of Hippo pathway activation, the present study demonstrates that inhibition of Yap1 activity has anti-tumor effects in gastric tumors driven by oncogenic mutations and inflammatory cytokines. We show that Yap1 is a key regulator of cell metabolism, proliferation, and immune responses in normal and neoplastic gastric epithelium. We propose that the Hippo pathway is targetable across gastric cancer subtypes and its therapeutic benefits are likely to be mediated by both cancer cell-intrinsic and -extrinsic mechanisms.
Collapse
Affiliation(s)
- Pathum Thilakasiri
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Ryan N O'Keefe
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Sarah Q To
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - David Chisanga
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Moritz F Eissmann
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Annalisa LE Carli
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Belinda Duscio
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - David Baloyan
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Rhynelle S Dmello
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - David Williams
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
- Department of Pathology, Austin Health, Heidelberg, Australia
| | - John Mariadason
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Ashleigh R Poh
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Bhupinder Pal
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Benjamin T Kile
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | | | - Kieran F Harvey
- Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Anatomy and Developmental Biology, and Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Michael Buchert
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Wei Shi
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Ashwini L Chand
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| |
Collapse
|
3
|
Long non-coding RNA SNHG1/microRNA-195-5p/Yes-associated protein axis affects the proliferation and metastasis of gastric cancer via the Hippo signaling pathway. Funct Integr Genomics 2022; 22:1043-1055. [PMID: 35819551 DOI: 10.1007/s10142-022-00876-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 06/10/2022] [Accepted: 06/10/2022] [Indexed: 01/18/2023]
Abstract
Long non-coding RNA (lncRNA) small nucleolar RNA host gene 1 (SNHG1) has been found to be highly expressed in gastric cancer (GC). However, the study for exploring the effects of SNHG1 and microRNA (miR)-195-5p on GC is limited. This research commits to unravel the regulatory effects of SNHG1, miRNA-195-5p, and Yes-associated protein 1 (YAP1) on GC. SNHG1, miR-195-5p and YAP1 levels in GC tissues and GC cells were detected. The GC cells were treated with various constructs altering SNHG1 or miR-195-5p expression to determine the biological activities of GC cell in vitro. The effect of SNHG1 inhibition on subcutaneous tumorigenesis of GC cells in a nude mouse model in vivo was detected. The binding relation among SNHG1, miR-195-5p, and YAP1 was validated. SNHG1 and YAP1 levels were elevated and miR-195-5p level was reduced in GC. Reduction of SNHG1 or elevation of miR-195-5p retarded GC cell biological activity in vitro. Downregulated SNHG1 suppressed tumor growth in vivo. SNHG1 bound to miR-195-5p, and miR-195-5p directly targeted YAP1. The downregulated SNHG1 hinders the biological behaviors of GC cells via the modulation of the miR-195-5p/YAP1 axis.
Collapse
|
4
|
Soltani R, Amini M, Mazaheri Moghaddam M, Jebelli A, Ahmadiyan S, Bidar N, Baradaran B, MotieGhader H, Asadi M, Mokhtarzadeh A. LncRNA DLGAP1-AS2 overexpression associates with gastric tumorigenesis: a promising diagnostic and therapeutic target. Mol Biol Rep 2022; 49:6817-6826. [PMID: 34981339 DOI: 10.1007/s11033-021-07038-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/29/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND Aberrant expression of long noncoding RNAs (lncRNAs) is associated with the progression of human cancers, including gastric cancer (GC). The function of lncRNA DLGAP1-AS2, as a promising oncogene, has been identified in several human cancers. Therefore, this study was aimed to explore the association of DLGAP1-AS2 with gastric tumorigenesis, as well. METHODS AND RESULTS The expression level of DLGAP1-AS2 was initially pre-evaluated in GC datasets from Gene Expression Omnibus (GEO). Moreover, qRT-PCR experiment was performed on 25 GC and 25 adjacent normal tissue samples. The Cancer Genome Atlas (TCGA) data were also analyzed for further validation. Consistent with data obtained from GEO datasets, qRT-PCR results revealed that DLGAP1-AS2 was significantly (p < 0.0032) upregulated in GC specimens compared to normal samples, which was additionally confirmed using TCGA analysis (p < 0.0001). DLGAP1-AS2 expression level was also correlated with age (p = 0.0008), lymphatic and vascular invasion (p = 0.0415) in internal samples as well as poor survival of GC patients (p = 0.00074) in GEO datasets. Also, Gene Ontology analysis illustrated that DLGAP1-AS2 may be involved in the cellular process, including hippo signaling, regulated by YAP1, as its valid downstream target, in GC samples. Moreover, ROC curve analysis showed the high accuracy of the DLGAP1-AS2 expression pattern as a diagnostic biomarker for GC. CONCLUSION Our findings indicated that DLGAP1-AS2 might display oncogenic properties through gastric tumorigenesis and could be suggested as a therapeutic, diagnostic, and prognostic target.
Collapse
Affiliation(s)
- Rogayeh Soltani
- Department of Biology, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Asiyeh Jebelli
- Department of Biology, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Sahar Ahmadiyan
- Department of Biology, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Negar Bidar
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Habib MotieGhader
- Department of Bioinformatics, Biotechnology Research Center, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Milad Asadi
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Turkey
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
5
|
m6A Methyltransferase 3 Promotes the Proliferation and Migration of Gastric Cancer Cells through the m6A Modification of YAP1. JOURNAL OF ONCOLOGY 2021; 2021:8875424. [PMID: 34394353 PMCID: PMC8357513 DOI: 10.1155/2021/8875424] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 06/28/2021] [Accepted: 07/24/2021] [Indexed: 12/14/2022]
Abstract
Gastric cancer is the most common gastrointestinal tumor with an increasing incidence. Furthermore, advanced gastric cancer is more common, but the mechanism underlying the proliferation and metastasis of gastric cancer has not been thoroughly explored. N6-methyladenosine (m6A) methyltransferase 3 (METTL3) may be involved in the proliferation and metastasis of gastric cancer. Therefore, Yes-associated protein 1 (YAP1) in the Hippo pathway was selected as the target, and the relationship between METTL3 and the proliferation and metastasis of gastric cancer was proved through a series of experiments. This research showed that the expression of m6A and METTL3 was upregulated in human gastric cancer tissues and gastric cancer cell lines. After lentiviral transfection, METTL3 silencing in AGS (human gastric adenocarcinoma cell line AGS) and MKN-45 (human gastric cancer cell line MKN-45) gastric cancer cell lines directly inhibited the proliferation, aggressiveness, and migration of gastric cancer cells. Mechanically, the inhibition of the YAP1-TEAD signaling pathway by peptide 17 reduces m6A methylation and the total mRNA level of YAP1. It also eliminates the promoting effect of METTL3 on the proliferation and migration of gastric cancer cells. In turn, the overexpression of YAP1 eliminates the inhibitory effect of METTL3 silencing on the proliferation, migration, and invasion of gastric cancer cells. This article proved that m6A methyltransferase METTL3 promoted the proliferation and migration of gastric cancer cells through the m6A modification of YAP1.
Collapse
|
6
|
Wu B, Tang X, Ke H, Zhou Q, Zhou Z, Tang S, Ke R. Gene Regulation Network of Prognostic Biomarker YAP1 in Human Cancers: An Integrated Bioinformatics Study. Pathol Oncol Res 2021; 27:1609768. [PMID: 34257617 PMCID: PMC8262238 DOI: 10.3389/pore.2021.1609768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/24/2021] [Indexed: 12/20/2022]
Abstract
Background: Yes-associated protein 1 (YAP1) is the main downstream effector of the Hippo signaling pathway, which is involved in tumorigenesis. This study aimed to comprehensively understand the prognostic performances of YAP1 expression and its potential mechanism in pan-cancers by mining databases. Methods: The YAP1 expression was evaluated by the Oncomine database and GEPIA tool. The clinical significance of YAP1 expression was analyzed by the UALCAN, GEPIA, and DriverDBv3 database. Then, the co-expressed genes with YAP1 were screened by the LinkedOmics, and annotated by the Metascape and DAVID database. Additionally, by the MitoMiner 4.0 v tool, the YAP1 co-expressed genes were screened to obtain the YAP1-associated mitochondrial genes that were further enriched by DAVID and analyzed by MCODE for the hub genes. Results: YAP1 was differentially expressed in human cancers. Higher YAP1 expression was significantly associated with poorer overall survival and disease-free survival in adrenocortical carcinoma (ACC), brain Lower Grade Glioma (LGG), and pancreatic adenocarcinoma (PAAD). The LinkedOmics analysis revealed 923 co-expressed genes with YAP1 in adrenocortical carcinoma, LGG and PAAD. The 923 genes mainly participated in mitochondrial functions including mitochondrial gene expression and mitochondrial respiratory chain complex I assembly. Of the 923 genes, 112 mitochondrial genes were identified by MitoMiner 4.0 v and significantly enriched in oxidative phosphorylation. The MCODE analysis identified three hub genes including CHCHD1, IDH3G and NDUFAF5. Conclusion: Our findings showed that the YAP1 overexpression could be a biomarker for poor prognosis in ACC, LGG and PAAD. Specifically, the YAP1 co-expression genes were mainly involved in the regulation of mitochondrial function especially in oxidative phosphorylation. Thus, our findings provided evidence of the carcinogenesis of YAP1 in human cancers and new insights into the mechanisms underlying the role of YAP1 in mitochondrial dysregulation.
Collapse
Affiliation(s)
- Baojin Wu
- Department of Plastic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinjie Tang
- Department of Plastic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Honglin Ke
- Department of Emergency, Huashan Hospital Affiliated to Fudan University, Shanghai, China
| | - Qiong Zhou
- Department of Statistics, Florida State University, Tallahassee, FL, United States
| | - Zhaoping Zhou
- Department of Plastic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Shao Tang
- Department of Statistics, Florida State University, Tallahassee, FL, United States
| | - Ronghu Ke
- Department of Plastic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Li Y, Sun R, Zhang Y, Yuan Y, Miao Y. A methylation-based mRNA signature predicts survival in patients with gastric cancer. Cancer Cell Int 2020; 20:284. [PMID: 32647495 PMCID: PMC7336496 DOI: 10.1186/s12935-020-01374-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/22/2020] [Indexed: 11/22/2022] Open
Abstract
Background Evidence suggests that altered DNA methylation plays a causative role in the occurrence, progression and prognosis of gastric cancer (GC). Thus, methylated-differentially expressed genes (MDEGs) could potentially serve as biomarkers and therapeutic targets in GC. Methods Four genomics profiling datasets were used to identify MDEGs. Gene Ontology enrichment and Kyoto Encyclopaedia of Genes and Genomes pathway enrichment analysis were used to explore the biological roles of MDEGs in GC. Univariate Cox and LASSO analysis were used to identify survival-related MDEGs and to construct a MDEGs-based signature. The prognostic performance was evaluated in two independent cohorts. Results We identified a total of 255 MDEGs, including 192 hypermethylation-low expression and 63 Hypomethylation-high expression genes. The univariate Cox regression analysis showed that 83 MDEGs were associated with overall survival. Further we constructed an eight-MDEGs signature that was independent predictive of prognosis in the training cohort. By applying the eight-MDEGs signature, patients in the training cohort could be categorized into high-risk or low-risk subgroup with significantly different overall survival (HR = 2.62, 95% CI 1.71–4.02, P < 0.0001). The prognostic value of the eight-MDEGs signature was confirmed in another independent GEO cohort (HR = 1.35, 95% CI 1.03–1.78, P = 0.0302) and TCGA-GC cohort (HR = 1.85, 95% CI 1.16–2.94, P = 0.0084). Multivariate cox regression analysis proved the eight-MDEGs signature was an independent prognostic factor for GC. Conclusion We have thus established an innovative eight-MDEGs signature that is predictive of overall survival and could be a potentially useful guide for personalized treatment of GC patients.
Collapse
Affiliation(s)
- Yang Li
- Department of Central Laboratory, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, 221009 China
| | - Rongrong Sun
- Department of Medical Oncology, Xuzhou Central Hospital, Clinical School of Xuzhou Medical University, Xuzhou, 221009 China
| | - Youwei Zhang
- Department of Medical Oncology, Xuzhou Central Hospital, Clinical School of Xuzhou Medical University, Xuzhou, 221009 China
| | - Yuan Yuan
- Department of Medical Oncology, Xuzhou Central Hospital, Clinical School of Xuzhou Medical University, Xuzhou, 221009 China
| | - Yufeng Miao
- Department of Medical Oncology, The First Peoples' Hospital of Wenling City, Wenling, 317500 China
| |
Collapse
|
8
|
TAZ Controls Helicobacter pylori-Induced Epithelial-Mesenchymal Transition and Cancer Stem Cell-Like Invasive and Tumorigenic Properties. Cells 2020; 9:cells9061462. [PMID: 32545795 PMCID: PMC7348942 DOI: 10.3390/cells9061462] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/01/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022] Open
Abstract
Helicobacter pylori infection, the main risk factor for gastric cancer (GC), leads to an epithelial–mesenchymal transition (EMT) of gastric epithelium contributing to gastric cancer stem cell (CSC) emergence. The Hippo pathway effectors yes-associated protein (YAP) and transcriptional co-activator with PDZ binding motif (TAZ) control cancer initiation and progression in many cancers including GC. Here, we investigated the role of TAZ in the early steps of H. pylori-mediated gastric carcinogenesis. TAZ implication in EMT, invasion, and CSC-related tumorigenic properties were evaluated in three gastric epithelial cell lines infected by H. pylori. We showed that H. pylori infection increased TAZ nuclear expression and transcriptional enhancer TEA domain (TEAD) transcription factors transcriptional activity. Nuclear TAZ and zinc finger E-box-binding homeobox 1 (ZEB1) were co-overexpressed in cells harboring a mesenchymal phenotype in vitro, and in areas of regenerative hyperplasia in gastric mucosa of H. pylori-infected patients and experimentally infected mice, as well as at the invasive front of gastric carcinoma. TAZ silencing reduced ZEB1 expression and EMT phenotype, and strongly inhibited invasion and tumorsphere formation induced by H. pylori. In conclusion, TAZ activation in response to H. pylori infection contributes to H. pylori-induced EMT, invasion, and CSC-like tumorigenic properties. TAZ overexpression in H. pylori-induced pre-neoplastic lesions and in GC could therefore constitute a biomarker of early transformation in gastric carcinogenesis.
Collapse
|
9
|
Li X, Li Y, Han Y, Dong B, Liu D, Che L, Liu Y, Wang Y. miR-205 Promotes Apoptosis of Cervical Cancer Cells and Enhances Drug Sensitivity of Cisplatin by Inhibiting YAP1. Cancer Biother Radiopharm 2020; 35:338-344. [PMID: 32379984 DOI: 10.1089/cbr.2019.2983] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective: Elevated expression of Yes-associated protein (YAP1) involves in the pathogenesis of cervical cancer. Bioinformatics analysis showed a targeting relationship between miR-205 and the 3'-UTR of YAP1. In this study, we aim to explore the role of miR-205 in the proliferation, apoptosis, or cisplatin (CDDP) resistance of cervical cancer cells. Patients and Methods: The dual luciferase reporter gene assay verified the relationship between miR-205 and YAP1. The CDDP-resistant cell line Hela/CDDP cells were cultured in vitro and divided into miR-NC group, miR-205 mimic group, and miR-205 inhibitor group followed by analysis of the expression of miR-205 and YAP1 mRNA by quantitative real-time polymerase chain reaction (qRT-PCR), and YAP1 protein level by western blot. Results: There was a targeted relationship between miR-205 and YAP1 mRNA. Compared with cervical cell line HCerEpiC cells, miR-205 expression was significantly decreased and YAP1 mRNA and protein expression was significantly increased in Hela cells (p < 0.01). Compared with miR-NC group, YAP1 protein expression in HeLa/CDDP cells was significantly decreased, cell apoptosis was increased, and proliferation was inhibited in miR-205 mimic-transfected Hela/CDDP cells (p < 0.01). Opposite results were obtained in miR-205 inhibitor-transfected Hela/CDDP cells. Conclusions: The expression of miR-205 is related to the CDDP resistance of cervical cancer cells. Increasing the expression of miR-205 can downregulate the expression of YAP1, inhibit the proliferation and promote apoptosis of cervical cancer cells, and enhance the sensitivity to CDDP.
Collapse
Affiliation(s)
- Xingmei Li
- Department of Gynecology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Yuewen Li
- Department of Gynecology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Yuning Han
- Department of Gynecology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Bing Dong
- Department of Gynecology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Dan Liu
- Department of Genetics, Qiqihar Medical University, Qiqihar, China
| | - Liqun Che
- Department of Endocrinology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Yu Liu
- Department of Gynecology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Yuchun Wang
- Department of Pharmacology, Qiqihar Medical University, Qiqihar, China
| |
Collapse
|
10
|
Kohmoto T, Masuda K, Shoda K, Takahashi R, Ujiro S, Tange S, Ichikawa D, Otsuji E, Imoto I. Claudin-6 is a single prognostic marker and functions as a tumor-promoting gene in a subgroup of intestinal type gastric cancer. Gastric Cancer 2020; 23:403-417. [PMID: 31654186 DOI: 10.1007/s10120-019-01014-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 10/12/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND We aimed to identify novel tumor-promoting drivers highly expressed in gastric cancer (GC) that contribute to worsened prognosis in affected patients. METHODS Genes whose expression was increased and correlated with worse prognosis in GC were screened using datasets from the Cancer Genome Atlas and Gene Expression Omnibus. We examined Claudin-6 (CLDN6) immunoreactivity in GC tissues and the effect of CLDN6 on cellular functions in GC cell lines. The mechanisms underlying GC-promoting function of CLDN6 were also investigated. RESULTS CLDN6 was identified as a gene overexpressed in GC tumors as compared with adjacent non-tumorous tissues and whose increased expression was positively correlated with worse overall survival of GC patients, particularly those with Lauren's intestinal type GC, in data from multiple publicly available datasets. Additionally, membranous CLDN6 immunoreactivity detected in intestinal type GC tumors was correlated with worse overall survival. In CLDN6-expressing GC cells, silencing of CLDN6 inhibited cell proliferation and migration/invasion abilities, possibly via suppressing transcription of YAP1 and its downstream transcriptional targets at least in part. CONCLUSIONS This study identified CLDN6 as a GC-promoting gene, suggesting that CLDN6 to be a possible single prognostic marker and promising therapeutic target for a subset of GC patients.
Collapse
Affiliation(s)
- Tomohiro Kohmoto
- Department of Human Genetics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Tokushima, 770-8503, Japan.,Division of Molecular Genetics, Aichi Cancer Center Research Institute, 1-1 Kanokoden Chikusa-ku, Nagoya, Aichi, 464-8681, Japan
| | - Kiyoshi Masuda
- Kawasaki Medical School, Kurashiki, Okayama, 701-0192, Japan
| | - Katsutoshi Shoda
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, 602-8566, Japan
| | - Rizu Takahashi
- Department of Human Genetics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Tokushima, 770-8503, Japan
| | - Sae Ujiro
- Department of Human Genetics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Tokushima, 770-8503, Japan
| | - Shoichiro Tange
- Department of Human Genetics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Tokushima, 770-8503, Japan
| | - Daisuke Ichikawa
- First Department of Surgery, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, 409-3898, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Kyoto, 602-8566, Japan
| | - Issei Imoto
- Department of Human Genetics, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Tokushima, 770-8503, Japan. .,Division of Molecular Genetics, Aichi Cancer Center Research Institute, 1-1 Kanokoden Chikusa-ku, Nagoya, Aichi, 464-8681, Japan. .,Department of Cancer Genetics, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan.
| |
Collapse
|
11
|
Molina-Castro SE, Tiffon C, Giraud J, Boeuf H, Sifre E, Giese A, Belleannée G, Lehours P, Bessède E, Mégraud F, Dubus P, Staedel C, Varon C. The Hippo Kinase LATS2 Controls Helicobacter pylori-Induced Epithelial-Mesenchymal Transition and Intestinal Metaplasia in Gastric Mucosa. Cell Mol Gastroenterol Hepatol 2019; 9:257-276. [PMID: 31669263 PMCID: PMC6957828 DOI: 10.1016/j.jcmgh.2019.10.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 10/17/2019] [Accepted: 10/18/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Gastric carcinoma is related mostly to CagA+-Helicobacter pylori infection, which disrupts the gastric mucosa turnover and elicits an epithelial-mesenchymal transition (EMT) and preneoplastic transdifferentiation. The tumor suppressor Hippo pathway controls stem cell homeostasis; its core, constituted by the large tumor suppressor 2 (LATS2) kinase and its substrate Yes-associated protein 1 (YAP1), was investigated in this context. METHODS Hippo, EMT, and intestinal metaplasia marker expression were investigated by transcriptomic and immunostaining analyses in human gastric AGS and MKN74 and nongastric immortalized RPE1 and HMLE epithelial cell lines challenged by H pylori, and on gastric tissues of infected patients and mice. LATS2 and YAP1 were silenced using small interfering RNAs. A transcriptional enhanced associated domain (TEAD) reporter assay was used. Cell proliferation and invasion were evaluated. RESULTS LATS2 and YAP1 appear co-overexpressed in the infected mucosa, especially in gastritis and intestinal metaplasia. H pylori via CagA stimulates LATS2 and YAP1 in a coordinated biphasic pattern, characterized by an early transient YAP1 nuclear accumulation and stimulated YAP1/TEAD transcription, followed by nuclear LATS2 up-regulation leading to YAP1 phosphorylation and targeting for degradation. LATS2 and YAP1 reciprocally positively regulate each other's expression. Loss-of-function experiments showed that LATS2 restricts H pylori-induced EMT marker expression, invasion, and intestinal metaplasia, supporting a role of LATS2 in maintaining the epithelial phenotype of gastric cells and constraining H pylori-induced preneoplastic changes. CONCLUSIONS H pylori infection engages a number of signaling cascades that alienate mucosa homeostasis, including the Hippo LATS2/YAP1/TEAD pathway. In the host-pathogen conflict, which generates an inflammatory environment and perturbations of the epithelial turnover and differentiation, Hippo signaling appears as a protective pathway, limiting the loss of gastric epithelial cell identity that precedes gastric carcinoma development.
Collapse
Affiliation(s)
- Silvia Elena Molina-Castro
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France,University of Costa Rica, San José, Costa Rica
| | - Camille Tiffon
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France
| | - Julie Giraud
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France
| | - Hélène Boeuf
- INSERM, UMR1026, Bioingénierie tissulaire (BioTis), University of Bordeaux, Bordeaux, France
| | - Elodie Sifre
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France
| | - Alban Giese
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France
| | | | - Philippe Lehours
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France,Centre Hospitalier Universitaire (CHU) de Bordeaux, Bordeaux, France
| | - Emilie Bessède
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France,Centre Hospitalier Universitaire (CHU) de Bordeaux, Bordeaux, France
| | - Francis Mégraud
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France,Centre Hospitalier Universitaire (CHU) de Bordeaux, Bordeaux, France
| | - Pierre Dubus
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France,Centre Hospitalier Universitaire (CHU) de Bordeaux, Bordeaux, France
| | - Cathy Staedel
- INSERM, UMR1212, University of Bordeaux, Bordeaux, France,Cathy Staedel, PhD, INSERM U1212, “ARN: Régulations naturelle et artificielle” (ARNA)-Unités Mixtes de Recherche (UMR) Centre national de la recherche scientifique (CNRS) 5320, University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux Cedex, France. fax: +33 5 57 57 10 15.
| | - Christine Varon
- INSERM, UMR1053, Bordeaux Research in Translational Oncology, BaRITOn, University of Bordeaux, Bordeaux, France,Correspondence Address correspondence to: Christine Varon, PhD, INSERM U1053 Bordeaux Research in Translational Oncology (BaRITOn), University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux Cedex, France. fax: +33 5 56 79 60 18.
| |
Collapse
|
12
|
Kim SH, Jin H, Meng RY, Kim DY, Liu YC, Chai OH, Park BH, Kim SM. Activating Hippo Pathway via Rassf1 by Ursolic Acid Suppresses the Tumorigenesis of Gastric Cancer. Int J Mol Sci 2019; 20:E4709. [PMID: 31547587 PMCID: PMC6801984 DOI: 10.3390/ijms20194709] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 12/16/2022] Open
Abstract
The Hippo pathway is often dysregulated in many carcinomas, which results in various stages of tumor progression. Ursolic acid (UA), a natural compound that exists in many herbal plants, is known to obstruct cancer progression and exerts anti-carcinogenic effect on a number of human cancers. In this study, we aimed to examine the biological mechanisms of action of UA through the Hippo pathway in gastric cancer cells. MTT assay showed a decreased viability of gastric cancer cells after treatment with UA. Following treatment with UA, colony numbers and the sizes of gastric cancer cells were significantly diminished and apoptosis was observed in SNU484 and SNU638 cells. The invasion and migration rates of gastric cancer cells were suppressed by UA in a dose-dependent manner. To further determine the gene expression patterns that are related to the effects of UA, a microarray analysis was performed. Gene ontology analysis revealed that several genes, such as the Hippo pathway upstream target gene, ras association domain family (RASSF1), and its downstream target genes (MST1, MST2, and LATS1) were significantly upregulated by UA, while the expression of YAP1 gene, together with oncogenes (FOXM1, KRAS, and BATF), were significantly decreased. Similar to the gene expression profiling results, the protein levels of RASSF1, MST1, MST2, LATS1, and p-YAP were increased, whereas those of CTGF were decreased by UA in gastric cancer cells. The p-YAP expression induced in gastric cancer cells by UA was reversed with RASSF1 silencing. In addition, the protein levels in the Hippo pathway were increased in the UA-treated xenograft tumor tissues as compared with that in the control tumor tissues; thus, UA significantly inhibited the tumorigenesis of gastric cancer in vivo in xenograft animals. Collectively, UA diminishes the proliferation and metastasis of gastric cancer via the regulation of Hippo pathway through Rassf1, which suggests that UA can be used as a potential chemopreventive and therapeutic agent for gastric cancer.
Collapse
Affiliation(s)
- Seong-Hun Kim
- Department of Internal Medicine, Chonbuk National University Medical School, Jeonju 54907, Korea.
| | - Hua Jin
- Department of Physiology, Chonbuk National University Medical School, Jeonju 54907, Korea.
| | - Ruo Yu Meng
- Department of Physiology, Chonbuk National University Medical School, Jeonju 54907, Korea.
| | - Da-Yeah Kim
- Department of Physiology, Chonbuk National University Medical School, Jeonju 54907, Korea.
| | - Yu Chuan Liu
- Department of Physiology, Chonbuk National University Medical School, Jeonju 54907, Korea.
| | - Ok Hee Chai
- Department of Anatomy and Institute for Medical Sciences, Chonbuk National University Medical School, Jeonju 54907, Korea.
| | - Byung Hyun Park
- Department of Biochemistry, Chonbuk National University Medical School, Jeonju 54907, Korea.
| | - Soo Mi Kim
- Department of Physiology, Chonbuk National University Medical School, Jeonju 54907, Korea.
- Research Institute of Clinical Medicine of Chonbuk National University, Jeonju 54907, Korea.
- Biomedical Research Institute of Chonbuk National University Hospital, Jeonju 54907, Korea.
| |
Collapse
|
13
|
Mechanotransduction and Cytoskeleton Remodeling Shaping YAP1 in Gastric Tumorigenesis. Int J Mol Sci 2019; 20:ijms20071576. [PMID: 30934860 PMCID: PMC6480114 DOI: 10.3390/ijms20071576] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/14/2019] [Accepted: 03/26/2019] [Indexed: 02/07/2023] Open
Abstract
The essential role of Hippo signaling pathway in cancer development has been elucidated by recent studies. In the gastrointestinal tissues, deregulation of the Hippo pathway is one of the most important driving events for tumorigenesis. It is widely known that Yes-associated protein 1 (YAP1) and WW domain that contain transcription regulator 1 (TAZ), two transcriptional co-activators with a PDZ-binding motif, function as critical effectors negatively regulated by the Hippo pathway. Previous studies indicate the involvement of YAP1/TAZ in mechanotransduction by crosstalking with the extracellular matrix (ECM) and the F-actin cytoskeleton associated signaling network. In gastric cancer (GC), YAP1/TAZ functions as an oncogene and transcriptionally promotes tumor formation by cooperating with TEAD transcription factors. Apart from the classic role of Hippo-YAP1 cascade, in this review, we summarize the current investigations to highlight the prominent role of YAP1/TAZ as a mechanical sensor and responder under mechanical stress and address its potential prognostic and therapeutic value in GC.
Collapse
|
14
|
Piao L, Wang F, Wang Y, Yang Z, Li Q, Cui L, Yu Q. miR-424-5p Regulates Hepatoma Cell Proliferation and Apoptosis. Cancer Biother Radiopharm 2019; 34:196-202. [PMID: 30676784 DOI: 10.1089/cbr.2018.2625] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Yes-associated protein (Yes-associated protein 1 [YAP1]) is an important oncogene that is related to the pathogenesis and progression of liver cancer. It was found that miR-424-5p expression was significantly decreased in liver cancer tissues, revealing its anticancer effect. Bioinformatic analysis demonstrated the targeted relationship between miR-424-5p and the 3' untranslated region of YAP1. This study investigated the role of miR-424-5p in regulating YAP1 expression and affecting hepatoma cell proliferation and apoptosis. MATERIALS AND METHODS Tumors and normal liver tissues adjacent to tumors were collected from patients to detect the expression of miR-424-5p and YAP1. A dual-luciferase reporter gene assay was adopted to explore the targeted regulation between miR-424-5p and YAP1. Liver cancer HCCLM3 and MHCC97-L cells and normal liver HL-7702 cells were cultured in vitro to compare expression levels of miR-424-5p and YAP1. HCCLM3 and MHCC97-L cells were divided into the miR-NC group and miR-424-5p mimic group. Cell apoptosis was detected by flow cytometry. Cell proliferation was determined by EdU staining. RESULTS Compared with normal liver tissue, miR-424-5p expression was significantly decreased, while YAP1 mRNA and protein levels were obviously upregulated in liver cancer tissues, which were related to the clinical stage. A negative correlation was found between miR-424-5p and YAP1 mRNA levels in liver cancer tissues. Dual-luciferase reporter gene assay confirmed the targeted relationship between miR-424-5p and YAP1. miR-424-5p expression in HCCLM3 and MHCC97-L cells decreased compared with L20 cells, which correlated with malignancy. YAP1 level in HCCLM3 and MHCC97-L cells was significantly enhanced, which correlated with malignancy. miR-424-5p mimic transfection significantly downregulated YAP1 expression in HCCLM3 and MHCC97-L cells, resulting in enhanced apoptosis and attenuated cell proliferation. CONCLUSIONS Decreased miR-424-5p expression and increased YAP1 expression are found in patients with liver cancer. Increased miR-424-5p can inhibit YAP1 expression, attenuate hepatoma cell proliferation, and induce cell apoptosis.
Collapse
Affiliation(s)
- Lianshu Piao
- Department of Gastroenterology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Fei Wang
- Department of Gastroenterology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Yanyan Wang
- Department of Gastroenterology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Zirong Yang
- Department of Gastroenterology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Qianwei Li
- Department of Gastroenterology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Lifeng Cui
- Department of Gastroenterology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Qinggong Yu
- Department of Gastroenterology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| |
Collapse
|
15
|
Burclaff J, Mills JC. Plasticity of differentiated cells in wound repair and tumorigenesis, part II: skin and intestine. Dis Model Mech 2018; 11:11/9/dmm035071. [PMID: 30171151 PMCID: PMC6177008 DOI: 10.1242/dmm.035071] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recent studies have identified and begun to characterize the roles of regenerative cellular plasticity in many organs. In Part I of our two-part Review, we discussed how cells reprogram following injury to the stomach and pancreas. We introduced the concept of a conserved cellular program, much like those governing division and death, which may allow mature cells to become regenerative. This program, paligenosis, is likely necessary to help organs repair the numerous injuries they face over the lifetime of an organism; however, we also postulated that rounds of paligenosis and redifferentiation may allow long-lived cells to accumulate and store oncogenic mutations, and could thereby contribute to tumorigenesis. We have termed the model wherein differentiated cells can store mutations and then unmask them upon cell cycle re-entry the ‘cyclical hit’ model of tumorigenesis. In the present Review (Part II), we discuss these concepts, and cell plasticity as a whole, in the skin and intestine. Although differentiation and repair are arguably more thoroughly studied in skin and intestine than in stomach and pancreas, it is less clear how mature skin and intestinal cells contribute to tumorigenesis. Moreover, we conclude our Review by discussing plasticity in all four organs, and look for conserved mechanisms and concepts that might help advance our knowledge of tumor formation and advance the development of therapies for treating or preventing cancers that might be shared across multiple organs. Summary: This final installment of a two-part Review discusses how cycles of dedifferentiation and redifferentiation can promote tumorigenesis in the skin and intestine, showing how plasticity in these continuously renewing tissues might contribute to tumorigenesis.
Collapse
Affiliation(s)
- Joseph Burclaff
- Division of Gastroenterology, Departments of Medicine, Pathology & Immunology, and Developmental Biology, Washington University, St Louis, MO 63110, USA
| | - Jason C Mills
- Division of Gastroenterology, Departments of Medicine, Pathology & Immunology, and Developmental Biology, Washington University, St Louis, MO 63110, USA
| |
Collapse
|
16
|
Burclaff J, Mills JC. Plasticity of differentiated cells in wound repair and tumorigenesis, part I: stomach and pancreas. Dis Model Mech 2018; 11:dmm033373. [PMID: 30037967 PMCID: PMC6078397 DOI: 10.1242/dmm.033373] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
For the last century or so, the mature, differentiated cells throughout the body have been regarded as largely inert with respect to their regenerative potential, yet recent research shows that they can become progenitor-like and re-enter the cell cycle. Indeed, we recently proposed that mature cells can become regenerative via a conserved set of molecular mechanisms ('paligenosis'), suggesting that a program for regeneration exists alongside programs for death (apoptosis) and division (mitosis). In two Reviews describing how emerging concepts of cellular plasticity are changing how the field views regeneration and tumorigenesis, we present the commonalities in the molecular and cellular features of plasticity at homeostasis and in response to injury in multiple organs. Here, in part 1, we discuss these advances in the stomach and pancreas. Understanding the extent of cell plasticity and uncovering its underlying mechanisms may help us refine important theories about the origin and progression of cancer, such as the cancer stem cell model, as well as the multi-hit model of tumorigenesis. Ultimately, we hope that the new concepts and perspectives on inherent cellular programs for regeneration and plasticity may open novel avenues for treating or preventing cancers.
Collapse
Affiliation(s)
- Joseph Burclaff
- Division of Gastroenterology, Departments of Medicine, Pathology and Immunology, and Developmental Biology, Washington University, St Louis, MO 63110, USA
| | - Jason C Mills
- Division of Gastroenterology, Departments of Medicine, Pathology and Immunology, and Developmental Biology, Washington University, St Louis, MO 63110, USA
| |
Collapse
|