1
|
Zhu N, Zhao Y, Yan W, Wei L, Sang Q, Li J, Liu B, Yu B. Characterization of alternative splicing events and prognostic signatures in gastric cancer. Cancer Cell Int 2024; 24:167. [PMID: 38734676 PMCID: PMC11088037 DOI: 10.1186/s12935-024-03348-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Accumulating evidences indicate that the specific alternative splicing (AS) events are linked to the occurrence and prognosis of gastric cancer (GC). Nevertheless, the impact of AS is still unclear and needed to further elucidation. METHODS The expression profile of GC and normal samples were downloaded from TCGA. AS events were achieved from SpliceSeq database. Cox regression together with LASSO analysis were employed to identify survival-associated AS events (SASEs) and calculate risk scores. PPI and pathway enrichment analysis were implemented to determine the function and pathways of these genes. Kaplan-Meier (K-M) analysis and Receiver Operating Characteristic Curves were used to evaluate the clinical significance of genes of SASEs. Q-PCR were applied to validate the hub genes on the survival prognosis in 47 GC samples. Drug sensitivity and immune cell infiltration analysis were conducted. RESULTS In total, 48 140 AS events in 10 610 genes from 361 GC and 31 normal samples were analyzed. Through univariate Cox regression, 855 SASEs in 763 genes were screened out. Further, these SASEs were analyzed by PPI and 17 hub genes were identified. Meanwhile, using Lasso and multivariate Cox regression analysis, 135 SASEs in 132 genes related to 7 AS forms were further screened and a GC prognostic model was constructed. K-M curves indicates that high-risk group has poorer prognosis. And the nomogram analysis on the basis of the multivariate Cox analysis was disclosed the interrelationships between 7 AS forms and clinical parameters in the model. Five key genes were then screened out by PPI analysis and Differential Expression Gene analysis based on TCGA and Combined-dataset, namely STAT3, RAD51B, SOCS2, POLE2 and TSR1. The expression levels of AS in STAT3, RAD51B, SOCS2, POLE2 and TSR1 were all significantly correlated with survival by qPCR verification. Nineteen drugs were sensitized to high-risk patients and eight immune cells showed significantly different infiltration between the STAD and normal groups. CONCLUSIONS In this research, the prognostic model constructed by SASEs can be applied to predict the prognosis of GC patients and the selected key genes are expected to become new biomarkers and therapeutical targets for GC treatment.
Collapse
Affiliation(s)
- Nan Zhu
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yupeng Zhao
- Gastroenterological Surgery, The affiliated Wuxi No. 2, People's Hospital of Nanjing Medical University, Wuxi, 200240, China
| | - Wenjing Yan
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lan Wei
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qingqing Sang
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jianfang Li
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Bingya Liu
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Beiqin Yu
- Department of General Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
2
|
Zhou Z, Li C, Wang Z, Haybaeck J, Zhang C. Cd44v6 acts as a directional responding factor in the process of transcoelomic metastasis from gastric carcinoma to Krukenberg tumor. Expert Rev Mol Diagn 2023; 23:583-588. [PMID: 37409376 DOI: 10.1080/14737159.2023.2223981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 06/06/2023] [Indexed: 07/07/2023]
Abstract
INTRODUCTION Due to the limited number of studies focusing on the optimal treatment of multiple Krukenberg tumor (KT)-gastric carcinoma (KT - GC), it is necessary to conduct large-scale studies to confirm the definite role of serum tumor markers in the diagnosis and prognosis of KT. Moreover, the clinical significance of variant 6 of CD44 (CD44v6) in transcoelomic metastasis should be considered. AREAS COVERED This review covers molecular pre-cancer diagnosis, gastric carcinoma metastasis, and anti-cancer treatments. Additionally, gastrointestinal cancer metastasis is a key area for improvement. EXPERT OPINION The detection of CD44v6 differs in the World Health Organization Classification of Gastric Adenocarcinoma, the Lauren Classification of Gastric Adenocarcinoma, and the anatomic location of gastric adenocarcinoma. The results were compared among the three groups. The mechanism of gastric adenocarcinoma metastasis still requires further elucidation. CD44v6 molecular detection helps clarify the pre-cancer diagnosis of KT before seeding. If subsequent studies confirm its role as a signaling molecule, it could pave the way for new research directions in clinical practice; however, additional academic confirmation is necessary.
Collapse
Affiliation(s)
- Ziqi Zhou
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, P.R. China
- Faculty of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, P.R. China
| | - Can Li
- Faculty of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, P.R. China
| | - Zhiyu Wang
- Faculty of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, P.R. China
| | - Johannes Haybaeck
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Innsbruck, Austria
- Diagnostic and Research Center for Molecular Biomedicine, Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Cuiwei Zhang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, P.R. China
| |
Collapse
|
3
|
Giraud J, Seeneevassen L, Rousseau B, Bouriez D, Sifré E, Giese A, Nguyen TL, Tiffon C, Lippi Y, Azzi-Martin L, Pannequin J, Ménard A, Bessède E, Staedel C, Mégraud F, Belleannée G, Lehours P, Gronnier C, Dubus P, Varon C. CD44v3 is a marker of invasive cancer stem cells driving metastasis in gastric carcinoma. Gastric Cancer 2023; 26:234-249. [PMID: 36528833 PMCID: PMC9950191 DOI: 10.1007/s10120-022-01357-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 11/27/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Cancer stem cells (CSCs) are at the origin of tumour initiation and progression in gastric adenocarcinoma (GC). However, markers of metastasis-initiating cells remain unidentified in GC. In this study, we characterized CD44 variants expressed in GC and evaluated the tumorigenic and metastatic properties of CD44v3+ cells and their clinical significance in GC patients. METHODS Using GC cell lines and patient-derived xenografts, we evaluated CD44+ and CD44v3+ GC cells molecular signature and their tumorigenic, chemoresistance, invasive and metastatic properties, and expression in patients-derived tissues. RESULTS CD44v3+ cells, which represented a subpopulation of CD44+ cells, were detected in advanced preneoplastic lesions and presented CSCs chemoresistance and tumorigenic properties in vitro and in vivo. Molecular and functional analyses revealed two subpopulations of gastric CSCs: CD44v3+ CSCs with an epithelial-mesenchymal transition (EMT)-like signature, and CD44+/v3- CSCs with an epithelial-like signature; both were tumorigenic but CD44v3+ cells showed higher invasive and metastatic properties in vivo. CD44v3+ cells detected in the primary tumours of GC patients were associated with a worse prognosis. CONCLUSION CD44v3 is a marker of a subpopulation of CSCs with metastatic properties in GC. The identification of metastasis-initiating cells in GC represents a major advance for further development of anti-metastatic therapeutic strategies.
Collapse
Affiliation(s)
- Julie Giraud
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France
| | - Lornella Seeneevassen
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France
| | - Benoit Rousseau
- Animal Facility, University of Bordeaux, 33076 Bordeaux, France
| | - Damien Bouriez
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France ,Department of Digestive Surgery, Haut-Lévêque Hospital, 33000 Bordeaux, France ,CHU Bordeaux, 33076 Bordeaux, France
| | - Elodie Sifré
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France
| | - Alban Giese
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France
| | - Tra Ly Nguyen
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France
| | - Camille Tiffon
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France
| | - Yannick Lippi
- Toxalim Research Centre in Food Toxicology, Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Lamia Azzi-Martin
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France
| | - Julie Pannequin
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Armelle Ménard
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France
| | - Emilie Bessède
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France
| | - Cathy Staedel
- INSERM U1212, ARNA, University of Bordeaux, 33076 Bordeaux, France
| | - Francis Mégraud
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France ,CHU Bordeaux, 33076 Bordeaux, France ,Centre National de Référence des Campylobacters et Helicobacters, Pellegrin Hospital, 33076 Bordeaux, France
| | - Geneviève Belleannée
- CHU Bordeaux, 33076 Bordeaux, France ,Department of Histology and Pathology, Haut-Lévêque Hospital, 33000 Bordeaux, France
| | - Philippe Lehours
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France ,CHU Bordeaux, 33076 Bordeaux, France ,Centre National de Référence des Campylobacters et Helicobacters, Pellegrin Hospital, 33076 Bordeaux, France
| | - Caroline Gronnier
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France ,Department of Digestive Surgery, Haut-Lévêque Hospital, 33000 Bordeaux, France ,CHU Bordeaux, 33076 Bordeaux, France
| | - Pierre Dubus
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France ,CHU Bordeaux, 33076 Bordeaux, France ,Department of Histology and Pathology, Haut-Lévêque Hospital, 33000 Bordeaux, France
| | - Christine Varon
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076, Bordeaux, France.
| |
Collapse
|
4
|
Hou W, Kong L, Hou Z, Ji H. CD44 is a prognostic biomarker and correlated with immune infiltrates in gastric cancer. BMC Med Genomics 2022; 15:225. [PMCID: PMC9620622 DOI: 10.1186/s12920-022-01383-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 10/25/2022] [Indexed: 11/10/2022] Open
Abstract
Objective Gastric carcinoma is the most common malignant tumour of the human digestive system worldwide. CD44 serves as a marker for several tumour stem cells, including gastric cancer. However, the prognostic value of CD44 and its correlation with immune infiltration in gastric cancer remain unclear. Methods The relative expression level of CD44 RNA in gastric cancer was analysed in the TCGA and GEPIA2 databases and validated in the GEO database. Differences in CD44 between gastric cancer cell lines and normal cells were detected by real-time PCR, and the HPA database was used to analyse the differential expression of CD44 protein in gastric cancer and normal tissues. The effect of CD44 on the proliferation and migration of gastric cancer cells was detected by CCK8 and transwell assays. UALCAN was used to analyse the relationship between CD44 expression and clinical parameters, and the Kaplan‒Meier Plotter was used to evaluate the prognostic value, including overall survival (OS), progression-free survival (PFS) and post-progression survival (PPS). The CD44 gene and protein interaction network was constructed by using the Linked Omics, GeneMANIA, STRING and DisGeNET databases. GO and KEGG analyses and GSEA of CD44 were performed by using R language. The correlation between CD44 and immune infiltration was explored by using the TIMER, CIBERSORT and GEPIA databases. Results CD44 is highly expressed in gastric cancer compared with normal tissues. Inhibition of proliferation and migration of gastric cancer cells after CD44 knockdown was observed. The UALCAN database showed that CD44 was independent of sex in gastric cancer but correlated with cancer stage and lymph node metastasis. Kaplan‒Meier Plotter online analysis showed that OS, PFS and PPS were prolonged in the CD44 low-expression group. GO and KEGG analyses and GSEA results showed that CD44 was mainly located in the endoplasmic reticulum and the extracellular matrix containing collagen, which was mainly involved in protein digestion and absorption. TIMER, CIBERSORT and GEPIA showed that CD44 was associated with infiltrating immune cells and thereby affected survival prognosis. Conclusion CD44 is highly expressed in gastric cancer and is an independent prognostic factor associated with immune invasion, which can be used as a candidate prognostic biomarker to determine the prognosis associated with gastric immune invasion.
Collapse
Affiliation(s)
- Weiyan Hou
- grid.413851.a0000 0000 8977 8425College of Basic Medicine, Chengde Medical University, Chengde, China
| | - Lingwei Kong
- grid.413851.a0000 0000 8977 8425Department of Orthopaedics, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Zhiping Hou
- grid.413851.a0000 0000 8977 8425Department of Pathology, Chengde Medical University, Shangerdaohezi Avenue, Chengde, 067000 Hebei China
| | - Hairu Ji
- grid.413851.a0000 0000 8977 8425Department of Pathology, Chengde Medical University, Shangerdaohezi Avenue, Chengde, 067000 Hebei China
| |
Collapse
|
5
|
Pereira C, Park JH, Campelos S, Gullo I, Lemos C, Solorzano L, Martins D, Gonçalves G, Leitão D, Lee HJ, Kong SH, André A, Borges C, Almeida D, Wälbhy C, Almeida R, Kim WH, Carneiro F, Yang HK, Almeida GM, Oliveira C. Comparison of East-Asia and West-Europe cohorts explains disparities in survival outcomes and highlights predictive biomarkers of early gastric cancer aggressiveness. Int J Cancer 2021; 150:868-880. [PMID: 34751446 DOI: 10.1002/ijc.33872] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/09/2021] [Accepted: 10/22/2021] [Indexed: 11/06/2022]
Abstract
Surgical resection with lymphadenectomy and peri-operative chemotherapy is the universal mainstay for curative treatment of gastric cancer (GC) patients with loco-regional disease. However, GC survival remains asymmetric in West- and East-world regions. We hypothesize this asymmetry derives from differential clinical management. Therefore, we collected chemo-naïve GC patients from Portugal and South-Korea to explore specific immunophenotypic profiles related to disease aggressiveness, and clinicopathological factors potentially explaining associated overall survival (OS) differences. Clinicopathological and survival data were collected from chemo-naïve surgical cohorts from Portugal (West-Europe cohort (WE-C); n=170) and South-Korea (East-Asia cohort (EA-C); n=367), and correlated with immunohistochemical expression profiles of E-cadherin and CD44v6 obtained from consecutive tissue microarrays sections. Survival analysis revealed a subset of 12.4% of WE-C patients, whose tumors concomitantly express E-cadherin_abnormal and CD44v6_very-high, displaying extremely poor OS, even at TNM stages I and II. These WE-C stages I and II patients were particularly aggressive compared to all other, invading deeper into the gastric wall (p=0.032) and more often permeating the vasculature (p=0.018) and nerves (p=0.009). A similar immunophenotypic profile was found in 11.9% of EA-C patients, but unrelated to survival. Stage I and II EA-C patients displaying both biomarkers also permeated more lymphatic vessels (p=0.003), promoting lymph node (LN) metastasis (p=0.019), being diagnosed on average 8-years earlier and submitted to more extensive LN dissection than WE-C. Concomitant E-cadherin_abnormal/CD44v6_very-high expression predicts aggressiveness and poor survival of stage I and II GC submitted to conservative lymphadenectomy.
Collapse
Affiliation(s)
- Carla Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal.,Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135, Porto, Portugal.,Doctoral Programme in Biomedicine, Faculty of Medicine of the University of Porto, 4200-319, Porto, Portugal
| | - Ji-Hyeon Park
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Sofia Campelos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal.,Department of Pathology, Ipatimup Diagnostics, Institute of Molecular Pathology and Immunology, University of Porto, 4200-135, Porto, Portugal
| | - Irene Gullo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal.,Department of Pathology, Centro Hospitalar Universitário de São João, 4200-319, Porto, Portugal.,Faculty of Medicine of the University of Porto, 4200-319, Porto, Portugal
| | - Carolina Lemos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal.,UniGENe, IBMC - Institute for Molecular and Cell Biology, 4200-135, Porto, Portugal.,ICBAS - Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313, Porto, Portugal
| | - Leslie Solorzano
- Center for Image Analysis, Dept. of IT and SciLifeLab, Uppsala University, SE-751 05, Uppsala, Sweden
| | - Diana Martins
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal.,Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135, Porto, Portugal.,Department of Biomedical Laboratory Sciences, ESTESC- Coimbra Health School, Polytechnic Institute of Coimbra, 3046-854, Coimbra, Portugal
| | - Gilza Gonçalves
- Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135, Porto, Portugal
| | - Dina Leitão
- Department of Pathology, Centro Hospitalar Universitário de São João, 4200-319, Porto, Portugal.,Faculty of Medicine of the University of Porto, 4200-319, Porto, Portugal
| | - Hyuk-Joon Lee
- Department of Surgery, Seoul National University Hospital, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Seong-Ho Kong
- Department of Surgery, Seoul National University Hospital, Seoul, Korea.,Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Ana André
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal.,Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135, Porto, Portugal
| | - Clara Borges
- Medical Oncology Department, Centro Hospitalar Universitário de São João, 4200-319, Porto, Portugal
| | - Daniela Almeida
- Medical Oncology Department, Centro Hospitalar Universitário de São João, 4200-319, Porto, Portugal
| | - Carolina Wälbhy
- Center for Image Analysis, Dept. of IT and SciLifeLab, Uppsala University, SE-751 05, Uppsala, Sweden
| | - Raquel Almeida
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal.,Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135, Porto, Portugal.,Department of Pathology, Centro Hospitalar Universitário de São João, 4200-319, Porto, Portugal.,Faculty of Sciences of the University of Porto, 4169-007, Porto, Portugal
| | - Woo Ho Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Fátima Carneiro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal.,Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135, Porto, Portugal.,Department of Pathology, Centro Hospitalar Universitário de São João, 4200-319, Porto, Portugal.,Faculty of Medicine of the University of Porto, 4200-319, Porto, Portugal
| | - Han-Kwang Yang
- Department of Surgery, Seoul National University Hospital, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Gabriela M Almeida
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal.,Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135, Porto, Portugal.,Faculty of Medicine of the University of Porto, 4200-319, Porto, Portugal
| | - Carla Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal.,Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135, Porto, Portugal.,Faculty of Medicine of the University of Porto, 4200-319, Porto, Portugal
| |
Collapse
|
6
|
Almeida GM, Pereira C, Park JH, Lemos C, Campelos S, Gullo I, Martins D, Gonçalves G, Leitão D, Neto JL, André A, Borges C, Almeida D, Lee HJ, Kong SH, Kim WH, Carneiro F, Almeida R, Yang HK, Oliveira C. CD44v6 High Membranous Expression Is a Predictive Marker of Therapy Response in Gastric Cancer Patients. Biomedicines 2021; 9:biomedicines9091249. [PMID: 34572441 PMCID: PMC8465138 DOI: 10.3390/biomedicines9091249] [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] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 01/27/2023] Open
Abstract
In gastric cancer (GC), biomarkers that define prognosis and predict treatment response remain scarce. We hypothesized that the extent of CD44v6 membranous tumor expression could predict prognosis and therapy response in GC patients. Two GC surgical cohorts, from Portugal and South Korea (n = 964), were characterized for the extension of CD44v6 membranous immuno-expression, clinicopathological features, patient survival, and therapy response. The value of CD44v6 expression in predicting response to treatment and its impact on prognosis was determined. High CD44v6 expression was associated with invasive features (perineural invasion and depth of invasion) in both cohorts and with worse survival in the Portuguese GC cohort (HR 1.461; 95% confidence interval 1.002–2.131). Patients with high CD44v6 tumor expression benefited from conventional chemotherapy in addition to surgery (p < 0.05), particularly those with heterogeneous CD44v6-positive and -negative populations (CD44v6_3+) (p < 0.007 and p < 0.009). Our study is the first to identify CD44v6 high membranous expression as a potential predictive marker of response to conventional treatment, but it does not clarify CD44v6 prognostic value in GC. Importantly, our data support selection of GC patients with high CD44v6-expressing tumors for conventional chemotherapy in addition to surgery. These findings will allow better stratification of GC patients for treatment, potentially improving their overall survival.
Collapse
Affiliation(s)
- Gabriela M Almeida
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- Ipatimup—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal;
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
| | - Carla Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- Ipatimup—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal;
- Doctoral Programme in Biomedicine, Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
| | - Ji-Hyeon Park
- Department of Surgery, Seoul National University Hospital, Seoul 03080, Korea; (J.-H.P.); (H.-J.L.); (S.-H.K.); (H.-K.Y.)
| | - Carolina Lemos
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- UnIGENe, IBMC—Institute for Molecular and Cell Biology, 4200-135 Porto, Portugal
- ICBAS—Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Sofia Campelos
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- Department of Pathology, Ipatimup Diagnostics, Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Irene Gullo
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
- Department of Pathology, Centro Hospitalar Universitário de São João, 4200-319 Porto, Portugal
| | - Diana Martins
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- Ipatimup—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal;
- Department of Biomedical Laboratory Sciences, ESTeSC—Coimbra Health School, Polytechnic Institute of Coimbra, 3046-854 Coimbra, Portugal
| | - Gilza Gonçalves
- Ipatimup—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal;
| | - Dina Leitão
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
- Department of Pathology, Centro Hospitalar Universitário de São João, 4200-319 Porto, Portugal
| | - João Luís Neto
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal;
| | - Ana André
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- Ipatimup—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal;
| | - Clara Borges
- Medical Oncology Department, Centro Hospitalar Universitário de São João, 4200-319 Porto, Portugal; (C.B.); (D.A.)
| | - Daniela Almeida
- Medical Oncology Department, Centro Hospitalar Universitário de São João, 4200-319 Porto, Portugal; (C.B.); (D.A.)
| | - Hyuk-Joon Lee
- Department of Surgery, Seoul National University Hospital, Seoul 03080, Korea; (J.-H.P.); (H.-J.L.); (S.-H.K.); (H.-K.Y.)
- Cancer Research Institute, Seoul National University, Seoul 03080, Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Seong-Ho Kong
- Department of Surgery, Seoul National University Hospital, Seoul 03080, Korea; (J.-H.P.); (H.-J.L.); (S.-H.K.); (H.-K.Y.)
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Woo Ho Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea;
| | - Fátima Carneiro
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- Ipatimup—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal;
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
- Department of Pathology, Centro Hospitalar Universitário de São João, 4200-319 Porto, Portugal
| | - Raquel Almeida
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- Ipatimup—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal;
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
- Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Han-Kwang Yang
- Department of Surgery, Seoul National University Hospital, Seoul 03080, Korea; (J.-H.P.); (H.-J.L.); (S.-H.K.); (H.-K.Y.)
- Cancer Research Institute, Seoul National University, Seoul 03080, Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Carla Oliveira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (G.M.A.); (C.P.); (C.L.); (S.C.); (I.G.); (D.M.); (A.A.); (F.C.); (R.A.)
- Ipatimup—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal;
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
- Correspondence: ; Tel.: +351-220-408-800
| |
Collapse
|
7
|
Liu T, Huang X, Zhao L, Xiao Z, Li Z, Xin Y, Yang S, Guo D, Zhao W, Mi Y, Li H. Distinguishable Targeting of Non-Small Cell Lung Cancer Using Hyaluronan Functionalized Platinum Nanoclusters and Their Inhibition Behaviors of Proliferation, Invasion, Migration. ChemistryOpen 2021; 10:882-888. [PMID: 34363352 PMCID: PMC8409085 DOI: 10.1002/open.202100070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/04/2021] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is the leading cause of cancer deaths worldwide and most cancer patients receiving conventional chemotherapy suffer from severe side effects due to the non-selective effects of chemotherapeutic drugs on normal cells. Targeted nanomaterials can obtain excellent accumulation at the tumor site through their active or passive targeting mechanisms, thereby reducing the toxicity of the drugs in various ways. In this study, hyaluronic acid (HA) which could specifically bind to CD44 on the surface of tumor cells, was used to modify amine-caged platinum nanoclusters (Pt NCs-NH2 ) to obtain targeting HA-Pt NCs-NH2 . Based on the differential expression of CD44 on the surface of three lung cells (non-small cell lung cancer cell H1299, small cell lung cancer cell H446, and embryonic lung fibroblast HFL1), HA-Pt NCs-NH2 can differentially enter the three cells and achieve their targeting of non-small cell lung cancer cell (NSCLC) cells. Pt NCs significantly inhibited the proliferation, migration and invasion of NSCLC cells and induced their apoptosis in comparison of classical cisplatin and carboplatin, showing a bright future in early diagnosis and treatment of NSCLC.
Collapse
Affiliation(s)
- Ting Liu
- Department of Respiratory and Critical Care MedicineThe Fifth Affiliation Hospital of Zhengzhou UniversityNo. 3 Kangfuqian StreetZhengzhou450052China
| | - Xin Huang
- School of TextilesZhongyuan University of TechnologyNo. 41 Zhongyuan Road (M)Zhengzhou450007China
| | - Lingyun Zhao
- Department of Respiratory and Critical Care MedicineThe Fifth Affiliation Hospital of Zhengzhou UniversityNo. 3 Kangfuqian StreetZhengzhou450052China
| | - Zhongqing Xiao
- Department of Respiratory and Critical Care MedicineThe Fifth Affiliation Hospital of Zhengzhou UniversityNo. 3 Kangfuqian StreetZhengzhou450052China
| | - Zengbei Li
- School of TextilesZhongyuan University of TechnologyNo. 41 Zhongyuan Road (M)Zhengzhou450007China
| | - Yi Xin
- Intensive Care UnitZhengzhou Orthopedics HospitalNo. 56 Longhai RoadZhengzhou450052China
| | - Shanshan Yang
- Department of Respiratory and Critical Care MedicineThe Fifth Affiliation Hospital of Zhengzhou UniversityNo. 3 Kangfuqian StreetZhengzhou450052China
| | - Di Guo
- Department of Respiratory and Critical Care MedicineThe Fifth Affiliation Hospital of Zhengzhou UniversityNo. 3 Kangfuqian StreetZhengzhou450052China
| | - Wenfei Zhao
- Department of Respiratory and Critical Care MedicineThe Fifth Affiliation Hospital of Zhengzhou UniversityNo. 3 Kangfuqian StreetZhengzhou450052China
| | - Yang Mi
- Henan Key Laboratory for Helicobacter pylori & Microbiota and GI CancerMarshall Medical Research CentreThe Fifth Affiliated Hospital of Zhengzhou UniversityNo. 3 Kangfuqian StreetZhengzhou450052China
| | - Hongyun Li
- Department of Respiratory and Critical Care MedicineThe Fifth Affiliation Hospital of Zhengzhou UniversityNo. 3 Kangfuqian StreetZhengzhou450052China
| |
Collapse
|
8
|
Hardas A, Suárez-Bonnet A, Beck S, Becker WE, Ramírez GA, Priestnall SL. Canine Gastric Carcinomas: A Histopathological and Immunohistochemical Study and Similarities with the Human Counterpart. Animals (Basel) 2021; 11:ani11051409. [PMID: 34069167 PMCID: PMC8156491 DOI: 10.3390/ani11051409] [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] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/19/2021] [Accepted: 05/10/2021] [Indexed: 01/06/2023] Open
Abstract
Simple Summary Gastric carcinoma (GC) continues to be one of the leading causes of death in humans and is the most common neoplasm in the stomachs of dogs. In both species, previous studies have demonstrated that the disease is heterogeneous, with genetic and environmental factors playing a quintessential role in disease pathogenesis. Compared to humans, the incidence of gastric carcinoma in dogs is low although, in a small number of breeds, a higher incidence has been reported. In dogs, the etiology and molecular pathways involved remain largely unknown. This retrospective study reviews current signalment data, evaluates the inflammatory component and association with Helicobacter spp. presence in various canine gastric carcinoma histological subtypes, and investigates potential molecular pathways involved in one of the largest study cohorts to date. The benefit of such a comparative study is to highlight the parallel histological features and molecular pathways between dogs and humans. Abstract Canine gastric carcinoma (CGC) affects both sexes in relatively equal proportions, with a mean age of nine years, and the highest frequency in Staffordshire bull terriers. The most common histological subtype in 149 CGC cases was the undifferentiated carcinoma. CGCs were associated with increased chronic inflammation parameters and a greater chronic inflammatory score when Helicobacter spp. were present. Understanding the molecular pathways of gastric carcinoma is challenging. All markers showed variable expression for each subtype. Expression of the cell cycle regulator 14-3-3σ was positive in undifferentiated, tubular and papillary carcinomas. This demonstrates that 14-3-3σ could serve as an immunohistochemical marker in routine diagnosis and that mucinous, papillary and signet-ring cell (SRC) carcinomas follow a 14-3-3σ independent pathway. p16, another cell cycle regulator, showed increased expression in mucinous and SRC carcinomas. Expression of the adhesion molecules E-cadherin and CD44 appear context-dependent, with switching within tumor emboli potentially playing an important role in tumor cell survival, during invasion and metastasis. Within neoplastic emboli, acinar structures lacked expression of all markers, suggesting an independent molecular pathway that requires further investigation. These findings demonstrate similarities and differences between dogs and humans, albeit further clinicopathological data and molecular analysis are required.
Collapse
Affiliation(s)
- Alexandros Hardas
- Department of Pathobiology & Population Sciences, The Royal Veterinary College, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK; (A.S.-B.); (W.E.B.); (S.L.P.)
- Correspondence:
| | - Alejandro Suárez-Bonnet
- Department of Pathobiology & Population Sciences, The Royal Veterinary College, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK; (A.S.-B.); (W.E.B.); (S.L.P.)
| | - Sam Beck
- VPG Histology, Horfield, Bristol BS7 0BJ, UK;
| | - William E. Becker
- Department of Pathobiology & Population Sciences, The Royal Veterinary College, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK; (A.S.-B.); (W.E.B.); (S.L.P.)
| | - Gustavo A. Ramírez
- Department of Animal Science, School of Agriculture, Food Science and Veterinary Medicine (ETSEA), University of Lleida, 25198 Lleida, Spain;
| | - Simon L. Priestnall
- Department of Pathobiology & Population Sciences, The Royal Veterinary College, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK; (A.S.-B.); (W.E.B.); (S.L.P.)
| |
Collapse
|
9
|
Solorzano L, Pereira C, Martins D, Almeida R, Carneiro F, Almeida GM, Oliveira C, Wahlby C. Towards Automatic Protein Co-Expression Quantification in Immunohistochemical TMA Slides. IEEE J Biomed Health Inform 2021; 25:393-402. [PMID: 32750958 DOI: 10.1109/jbhi.2020.3008821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Immunohistochemical (IHC) analysis of tissue biopsies is currently used for clinical screening of solid cancers to assess protein expression. The large amount of image data produced from these tissue samples requires specialized computational pathology methods to perform integrative analysis. Even though proteins are traditionally studied independently, the study of protein co-expression may offer new insights towards patients' clinical and therapeutic decisions. To explore protein co-expression, we constructed a modular image analysis pipeline to spatially align tissue microarray (TMA) image slides, evaluate alignment quality, define tumor regions, and ultimately quantify protein expression, before and after tumor segmentation. The pipeline was built with open-source tools that can manage gigapixel slides. To evaluate the consensus between pathologist and computer, we characterized a cohort of 142 gastric cancer (GC) cases regarding the extent of E-cadherin and CD44v6 expression. We performed IHC analysis in consecutive TMA slides and compared the automated quantification with the pathologists' manual assessment. Our results show that automated quantification within tumor regions improves agreement with the pathologists' classification. A co-expression map was created to identify the cores co-expressing both proteins. The proposed pipeline provides not only computational tools forwarding current pathology practices to explore co-expression, but also a framework for merging data and transferring information in learning-based approaches to pathology.
Collapse
|
10
|
Lourenço BN, Pereira RF, Barrias CC, Fischbach C, Oliveira C, Granja PL. Engineering Modular Half-Antibody Conjugated Nanoparticles for Targeting CD44v6-Expressing Cancer Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:295. [PMID: 33498669 PMCID: PMC7912417 DOI: 10.3390/nano11020295] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/17/2021] [Accepted: 01/19/2021] [Indexed: 02/06/2023]
Abstract
Gastric cancer (GC) remains a major cause of death worldwide mainly because of the late detection in advanced stage. Recently, we proposed CD44v6 as a relevant marker for early detection of GC, opening new avenues for GC-targeted theranostics. Here, we designed a modular nanoscale system that selectively targets CD44v6-expressing GC cells by the site-oriented conjugation of a new-engineered CD44v6 half-antibody fragment to maleimide-modified polystyrene nanoparticles (PNPs) via an efficient bioorthogonal thiol-Michael addition click chemistry. PNPs with optimal particle size (200 nm) for crossing a developed biomimetic CD44v6-associated GC stromal model were further modified with a heterobifunctional maleimide crosslinker and click conjugated to the novel CD44v6 half-antibody fragment, obtained by chemical reduction of full antibody, without affecting its bioactivity. Collectively, our results confirmed the specific targeting ability of CD44v6-PNPs to CD44v6-expressing cells (1.65-fold higher than controls), highlighting the potential of CD44v6 half-antibody conjugated nanoparticles as promising and clinically relevant tools for the early diagnosis and therapy of GC. Additionally, the rational design of our nanoscale system may be explored for the development of several other nanotechnology-based disease-targeted approaches.
Collapse
Affiliation(s)
- Bianca N. Lourenço
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (R.F.P.); (C.C.B.); (C.O.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- FEUP—Faculdade de Engenharia da Universidade do Porto, 4200-465 Porto, Portugal
| | - Rúben F. Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (R.F.P.); (C.C.B.); (C.O.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Cristina C. Barrias
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (R.F.P.); (C.C.B.); (C.O.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA;
- Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY 14853, USA
| | - Carla Oliveira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (R.F.P.); (C.C.B.); (C.O.)
- IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Departamento de Patologia, Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal
| | - Pedro L. Granja
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (R.F.P.); (C.C.B.); (C.O.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| |
Collapse
|
11
|
Zhang D, Huang J, Li W, Zhang Z, Zhu M, Feng Y, Zhao Y, Li Y, Lu S, He S. Screening and identification of a CD44v6 specific peptide using improved phage display for gastric cancer targeting. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1442. [PMID: 33313187 PMCID: PMC7723568 DOI: 10.21037/atm-19-4781] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Peptide probes can be applied for biomarker targeting to improve the diagnostic accuracy. Cluster of differentiation 44 (CD44) is up-regulated in gastric cancer (GC). Among all the variants of CD44, CD44v6 is reported the most promising biomarker for GC. The purpose of this study was generating and identification a peptide ligand specific to CD44v6. Methods A 12-mer phage peptide library was screened on CD44v overexpressed HEK-293 cells with an improved subtractive method. Five candidate sequences emerged. Candidate phages were selected using enzyme-linked immunosorbent assay and competitive inhibition assays. Then the sequence (designated ELT) was chosen for further study. Its binding affinity and specificity were verified on recombinant protein, GC cells, GC tissues and xenograft models based on BALB/c-nu/nu mice using dissociation constant calculation, immunofluorescence, immunohistochemistry and in vivo imaging separately. Results The dissociation constant of ELT with recombinant protein was 611.2 nM. ELT stained CD44v overexpressed HEK-293 but not the cell expressing wild-type CD44s. On GC cell lines, ELT co-stained with anti-CD44v6 antibody. ELT binding on tumor tissues significantly increased compared with that of paracancer tissues, also showed a linear positive correlation with CD44v6 expression. ELT specifically accumulated in tumor and eliminated in short time in vivo. Conclusions ELT can target GC in vitro and in vivo via CD44v6, indicating its potential to serve as a probe for GC targeting diagnosis and therapy.
Collapse
Affiliation(s)
- Dan Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Huang
- School of Electronic Information and Artificial Intelligence, Shannxi University of Science &Technology, Xi'an, China.,School of Materials Science and Engineering, Shannxi Normal University, Xi'an, China
| | - Weiming Li
- Department of Vascular Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhiyong Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meng Zhu
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yun Feng
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Zhao
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yarui Li
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shaoying Lu
- Department of Vascular Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shuixiang He
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
12
|
Alzeeb G, Metges JP, Corcos L, Le Jossic-Corcos C. Three-Dimensional Culture Systems in Gastric Cancer Research. Cancers (Basel) 2020; 12:E2800. [PMID: 33003476 PMCID: PMC7601358 DOI: 10.3390/cancers12102800] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/25/2020] [Accepted: 09/27/2020] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer (GC), which includes cancer of the esophagus, the oesophagogastric junction, and the stomach fundus, is highly deadly with strong regional influence, Asia being the most affected. GC is often detected at late stages, with 30% of metastatic cases at diagnosis. Many authors have devised models to both unravel the mechanisms of GC development and to evaluate candidate therapeutics. Among these models, 2D-cell cultures are progressively replaced by 3D-cell cultures that recapitulate, much more comprehensively, tumor cellular and genetic heterogeneity, as well as responsiveness to environmental changes, such as exposure to drugs or irradiation. With respect to the specifics of GC, there are high hopes from such model systems, especially with the aim of identifying prognostic markers and novel drug targets.
Collapse
Affiliation(s)
- George Alzeeb
- Inserm, University Brest, EFS, UMR 1078, GGB, F-29200 Brest, France; (G.A.); (L.C.)
| | - Jean-Philippe Metges
- CHU de Brest, Inserm, University Brest, EFS, UMR 1078, GGB, F-29200 Brest, France;
| | - Laurent Corcos
- Inserm, University Brest, EFS, UMR 1078, GGB, F-29200 Brest, France; (G.A.); (L.C.)
- CHU de Brest, Inserm, University Brest, EFS, UMR 1078, GGB, F-29200 Brest, France;
| | | |
Collapse
|
13
|
Zong Z, Li H, Ning Z, Hu C, Tang F, Zhu X, Tian H, Zhou T, Wang H. Integrative bioinformatics analysis of prognostic alternative splicing signatures in gastric cancer. J Gastrointest Oncol 2020; 11:685-694. [PMID: 32953152 DOI: 10.21037/jgo-20-117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background The potential prognostic value of alternative splicing (AS) variants and regulatory splicing factors in gastric carcinogenesis is unclear. We aimed to exploit the aberrant AS signatures and splicing factors involved in gastric cancer (GC) and to determine their prognostic predictive values. Methods We performed detailed data acquisition using the Cancer Genome Atlas project and profiled genome-wide AS signatures in a cohort of 190 patients with stomach adenocarcinoma (STAD). Prognostic prediction models and splicing correlation networks were assessed using an integrative bioinformatics analysis approach. Results We detected 1,308 overall survival (OS)-related AS signatures in 993 genes, most of which were favorable prognostic factors. Six splicing factors have been suggested to be dysregulated in GC, i.e., DHX15, PPP4R2, PRPF38B, RBM9, RBM15, and ILF3. Another notable finding was that most favorable prognosis AS events were positively correlated with expression of splicing factors, while a majority of poor survival prognostic AS genes were negatively associated with the expression of splicing factors. Conclusions To our knowledge, the current study provided the first comprehensive profiling of global modifications in the RNA splicing to identify survival associated AS signatures of GC specific genes. Our findings contribute to a better understanding of aberrant AS signatures and splicing factors in STAD, which can potentially be used as prognostic biomarkers and therapeutic targets for GC.
Collapse
Affiliation(s)
- Zhen Zong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hui Li
- Department of Rheumatology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhikun Ning
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Cegui Hu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fuxin Tang
- Department of Gastroenterological Surgery and Hernia Center, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaojian Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Huakai Tian
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Taicheng Zhou
- Department of Gastroenterological Surgery and Hernia Center, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - He Wang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
14
|
Feng H, Jin Z, Liu K, Peng Y, Jiang S, Wang C, Hu J, Shen X, Qiu W, Cheng X, Zhao R. Identification and validation of critical alternative splicing events and splicing factors in gastric cancer progression. J Cell Mol Med 2020; 24:12667-12680. [PMID: 32939931 PMCID: PMC7686978 DOI: 10.1111/jcmm.15835] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 08/12/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022] Open
Abstract
Gene expression and alternative splicing (AS) interact in complex ways to regulate biological process which is associated with cancer development. Here, by integrated analysis of gene expression and AS events, we aimed to identify the hub AS events and splicing factors relevant in gastric cancer development (GC). RNA‐seq data, clinical data and AS events of 348 GC samples were obtained from the TCGA and TCGASpliceSeq databases. Cox univariable and multivariable analyses, KEGG and GO pathway analyses were performed to identify hub AS events and splicing factor/spliceosome genes, which were further validated in 53 GCs. By bioinformatics methods, we found that gene AS event‐ and gene expression‐mediated GC progression shared the same mechanisms, such as PI3K/AKT pathway, but the involved genes were different. Though expression of 17 hub AS events were confirmed in 53 GC tissues, only 10 AS events in seven genes were identified as critical candidates related to GC progression, notably the AS events (Exon Skip) in CLSTN1 and SEC16A. Expression of these AS events in GC correlated with activation of the PI3K/AKT pathway. Genes with AS events associated with clinical parameters and prognosis were different from the genes whose mRNA levels were related to clinical parameters and prognosis. Besides, we further revealed that QKI and NOVA1 were the crucial splicing factors regulating expression of AS events in GC, but not spliceosome genes. Our integrated analysis revealed hub AS events in GC development, which might be the potential therapeutic targets for GC.
Collapse
Affiliation(s)
- Haoran Feng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of General Surgery, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhijian Jin
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kun Liu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of General Surgery, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Peng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of General Surgery, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Songyao Jiang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of General Surgery, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changgang Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of General Surgery, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiele Hu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of General Surgery, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyun Shen
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of General Surgery, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weihua Qiu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xi Cheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ren Zhao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of General Surgery, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Skipping Exon-v6 from CD44v6-Containing Isoforms Influences Chemotherapy Response and Self-Renewal Capacity of Gastric Cancer Cells. Cancers (Basel) 2020; 12:cancers12092378. [PMID: 32842638 PMCID: PMC7564355 DOI: 10.3390/cancers12092378] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/13/2020] [Accepted: 08/18/2020] [Indexed: 12/12/2022] Open
Abstract
De novo expressed CD44 isoforms containing exon-v6 are frequently associated with gastric cancer (GC) aggressiveness, and may predict chemotherapy response in vitro. Whether exon-v6 itself is responsible for conferring these properties to CD44v6-containing isoforms remains to be elucidated. CRISPR/Cas9 and Phosphorodiamidate Morpholino oligomers (PMOs) were used to induce specific exon-v6 skipping, maintaining the CD44 reading frame, in two GC cell lines endogenously expressing CD44v6. Cisplatin and 5-fluorouracil treatment response, and self-renewal ability was compared between CRISPR/Cas9-edited, CD44v6 knockdown and mock cells. We obtained homozygous genome-edited cell lines with exon-v6 deletion. Edited cells transcribed CD44v isoforms presenting in frame v5–v7 splicing, mimicking exon-v6 skipping. Results showed that removing specifically exon-v6 sensitizes cells to cisplatin and impairs cells’ self-renewal ability, similarly to CD44v6 knockdown. In parallel, we also tested a clinically feasible approach for transient exon-v6 skipping with a PMO-based strategy. We demonstrate that exon-v6 specific removal from CD44v isoforms increases cell sensitivity to cisplatin and impairs GC cells self-renewal. We trust that a PMO approach designed towards CD44v6 overexpressing GC cells may be a suitable approach to sensitize tumor cells for conventional therapy.
Collapse
|
16
|
Hu MN, Hu SH, Zhang XW, Xiong SM, Deng H. Overview on new progress of hereditary diffuse gastric cancer with CDH1 variants. TUMORI JOURNAL 2020; 106:346-355. [PMID: 32811340 DOI: 10.1177/0300891620949668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Hereditary diffuse gastric cancer (HDGC), comprising 1%-3% of gastric malignances, has been associated with CDH1 variants. Accumulating evidence has demonstrated more than 100 germline CDH1 variant types. E-cadherin encoded by the CDH1 gene serves as a tumor suppressor protein. CDH1 promoter hypermethylation and other molecular mechanisms resulting in E-cadherin dysfunction are involved in the tumorigenesis of HDGC. Histopathology exhibits characteristic signet ring cells, and immunohistochemical staining may show negativity for E-cadherin and other signaling proteins. Early HDGC is difficult to detect by endoscopy due to the development of lesions beneath the mucosa. Prophylactic gastrectomy is the most recommended treatment for pathogenic CDH1 variant carriers. Recent studies have promoted the progression of promising molecular-targeted therapies and management strategies. This review summarizes recent advances in CDH1 variant types, tumorigenesis mechanisms, diagnosis, and therapy, as well as clinical implications for future gene therapies.
Collapse
Affiliation(s)
- Mu-Ni Hu
- Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Shu-Hui Hu
- Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Xing-Wei Zhang
- Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Shu-Min Xiong
- Department of Ophthalmology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Huan Deng
- Molecular Medicine and Genetics Center, the Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China.,Renmin Institute of Forensic Medicine in Jiangxi, Nanchang, Jiangxi Province, China
| |
Collapse
|
17
|
Baião A, Sousa F, Oliveira AV, Oliveira C, Sarmento B. Effective intracellular delivery of bevacizumab via PEGylated polymeric nanoparticles targeting the CD44v6 receptor in colon cancer cells. Biomater Sci 2020; 8:3720-3729. [PMID: 32500879 DOI: 10.1039/d0bm00556h] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Colorectal cancer (CRC) is one of the most common and deadly cancers in the world, mainly due to its metastatic and metabolic ability. The CD44 receptor isoform containing exon 6 (CD44v6) is a transmembrane protein that plays an important role in the establishment of tumors and metastasis, which make this molecule a potential target for therapy and diagnosis of tumors. Aiming at a targeted therapy, the anti-VEGF monoclonal antibody (mAb) bevacizumab was loaded into poly(lactic-co-glycolic acid)-polyethylene glycol (PLGA-PEG) nanoparticles (NPs) functionalized with an antibody fragment (Fab) specific for CD44v6-expressing human cancer cells. The sizes of NPs were in the range of 150-250 nm and they had a negative charge between -5 and -10 mV, with an association efficiency (AE) of bevacizumab of 86%. v6 Fab-PLGA-PEG NPs containing bevacizumab specifically bonded to the CD44v6 cell surface receptor and exhibited higher internalization into CD44v6+ epithelial cells than bare and (-) Fab-PLGA-PEG NPs. To understand the biological effect of NP targeting, the intracellular levels of bevacizumab and VEGF were evaluated after the incubation of targeted and untargeted NPs. The intracellular levels of bevacizumab were significantly higher in cells incubated with v6 Fab-PLGA-PEG NPs and these NPs resulted in a significant decrease in the intracellular VEGF compared to untargeted NPs and free bevacizumab. PLGA-PEG NPs, surface-functionalized with a v6-specific Fab, have the potential to intracellularly deliver bevacizumab into CD44v6 expressing cancer cells.
Collapse
Affiliation(s)
- Ana Baião
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-393 Porto, Portugal.
| | | | | | | | | |
Collapse
|
18
|
Pereira C, Ferreira D, Mendes N, Granja PL, Almeida GM, Oliveira C. Expression of CD44v6-Containing Isoforms Influences Cisplatin Response in Gastric Cancer Cells. Cancers (Basel) 2020; 12:cancers12040858. [PMID: 32252293 PMCID: PMC7226224 DOI: 10.3390/cancers12040858] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/22/2020] [Accepted: 03/31/2020] [Indexed: 12/17/2022] Open
Abstract
CD44v6-containing isoforms are frequently de novo expressed in gastric cancer (GC). Whether CD44v6 has a central role in GC transformation and/or progression, whether it conditions response to therapy or whether it is only a bystander marker is still not known. Therefore, we aimed to clarify the role of CD44v6 in GC. We generated GC isogenic cell lines stably expressing CD44s or CD44v6 and tested them for different cancer hallmarks and response to cisplatin, and we further confirmed our findings in cells that endogenously express CD44v6. No correlation between overexpression of CD44v6 and the tested cancer hallmarks was observed, suggesting CD44v6 is not a driver of GC progression. Upon cisplatin treatment, CD44v6+ cells survive better and have lower apoptosis levels than CD44v6− cells, possibly due to concomitant activation of STAT3 and P38. In co-culture experiments, we discovered that CD44v6+ cells are involved in GC cell overgrowth after cisplatin treatment. In conclusion, we show that CD44v6 expression increases cell survival in response to cisplatin treatment in GC cells and that these cells override CD44v6-negative cells after cisplatin-treatment. This suggests that tumor expression of CD44v6-containing variants may condition the outcome of GC patients treated with chemotherapy.
Collapse
Affiliation(s)
- Carla Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.P.); (D.F.); (N.M.); (P.L.G.)
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Daniel Ferreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.P.); (D.F.); (N.M.); (P.L.G.)
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-180 Porto, Portugal
| | - Nuno Mendes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.P.); (D.F.); (N.M.); (P.L.G.)
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Pedro L. Granja
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.P.); (D.F.); (N.M.); (P.L.G.)
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-180 Porto, Portugal
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Gabriela M. Almeida
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.P.); (D.F.); (N.M.); (P.L.G.)
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- FMUP - Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
- Correspondence: (G.M.A.); (C.O.); Tel.: +351-225-570-785 (C.O.)
| | - Carla Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.P.); (D.F.); (N.M.); (P.L.G.)
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- FMUP - Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
- Correspondence: (G.M.A.); (C.O.); Tel.: +351-225-570-785 (C.O.)
| |
Collapse
|
19
|
Moreira IB, Pinto F, Gomes C, Campos D, Reis CA. Impact of Truncated O-glycans in Gastric-Cancer-Associated CD44v9 Detection. Cells 2020; 9:cells9020264. [PMID: 31973075 PMCID: PMC7072479 DOI: 10.3390/cells9020264] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 02/06/2023] Open
Abstract
CD44 variant isoforms are often upregulated in cancer and associated with increased aggressive tumor phenotypes. The CD44v9 is one of the major protein splice variant isoforms expressed in human gastrointestinal cancer cells. Immunodetection of CD44 isoforms like CD44v9 in tumor tissue is almost exclusively performed by using specific monoclonal antibodies. However, the structural variability conferred by both the alternative splicing and CD44 protein glycosylation is disregarded. In the present work, we have evaluated the role of O-glycosylation using glycoengineered gastric cancer models in the detection of CD44v9 by monoclonal antibodies. We demonstrated, using different technical approaches, that the presence of immature O-glycan structures, such as Tn and STn, enhance CD44v9 protein detection. These findings can have significant implications in clinical applications mainly at the detection and targeting of this cancer-related CD44v9 isoform and highlight the utmost importance of considering glycan structures in cancer biomarker detection and in therapy targeting.
Collapse
Affiliation(s)
- Inês B. Moreira
- I3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (I.B.M.); (F.P.); (C.G.)
- IPATIMUP–Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Filipe Pinto
- I3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (I.B.M.); (F.P.); (C.G.)
- IPATIMUP–Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Catarina Gomes
- I3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (I.B.M.); (F.P.); (C.G.)
- IPATIMUP–Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Diana Campos
- I3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (I.B.M.); (F.P.); (C.G.)
- IPATIMUP–Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- Correspondence: (D.C.); (C.A.R.)
| | - Celso A. Reis
- I3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (I.B.M.); (F.P.); (C.G.)
- IPATIMUP–Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4050-313 Porto, Portugal
- Correspondence: (D.C.); (C.A.R.)
| |
Collapse
|
20
|
Peng WZ, Liu JX, Li CF, Ma R, Jie JZ. hnRNPK promotes gastric tumorigenesis through regulating CD44E alternative splicing. Cancer Cell Int 2019; 19:335. [PMID: 31857793 PMCID: PMC6909542 DOI: 10.1186/s12935-019-1020-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/08/2019] [Indexed: 11/10/2022] Open
Abstract
Background The high prevalence of alternative splicing among genes implies the importance of genomic complexity in regulating normal physiological processes and diseases such as gastric cancer (GC). The standard form of stem cell marker CD44 (CD44S) and its alternatives with additional exons are reported to play important roles in multiple types of tumors, but the regulation mechanism of CD44 alternative splicing is not fully understood. Methods Here the expression of hnRNPK was analyzed among the Cancer Genome Atlas (TCGA) cohort of GC. The function of hnRNPK in GC cells was analyzed and its downstream targeted gene was identified by chromatin immunoprecipitation and dual luciferase report assay. Finally, effect of hnRNPK and its downstream splicing regulator on CD44 alternative splicing was investigated. Results The expression of hnRNPK was significantly increased in GC and its upregulation was associated with tumor stage and metastasis. Loss-of-function studies found that hnRNPK could promote GC cell proliferation, migration, and invasion. The upregulation of hnRNPK activates the expression of the splicing regulator SRSF1 by binding to the first motif upstream the start codon (- 65 to - 77 site), thereby increasing splicing activity and expression of an oncogenic CD44 isoform, CD44E (has additional variant exons 8 to 10, CD44v8-v10). Conclusion These findings revealed the importance of the hnRNPK-SRSF1-CD44E axis in promoting gastric tumorigenesis.
Collapse
Affiliation(s)
- Wei-Zhao Peng
- 1Department of General Surgery, China-Japan Friendship Hospital, Beijing, 100029 China
| | - Ji-Xi Liu
- 2Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, 100029 China
| | - Chao-Feng Li
- 1Department of General Surgery, China-Japan Friendship Hospital, Beijing, 100029 China
| | - Ren Ma
- 1Department of General Surgery, China-Japan Friendship Hospital, Beijing, 100029 China
| | - Jian-Zheng Jie
- 1Department of General Surgery, China-Japan Friendship Hospital, Beijing, 100029 China
| |
Collapse
|
21
|
Mereiter S, Martins ÁM, Gomes C, Balmaña M, Macedo JA, Polom K, Roviello F, Magalhães A, Reis CA. O‐glycan truncation enhances cancer‐related functions of
CD
44 in gastric cancer. FEBS Lett 2019; 593:1675-1689. [DOI: 10.1002/1873-3468.13432] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 05/04/2019] [Accepted: 05/06/2019] [Indexed: 01/16/2023]
Affiliation(s)
- Stefan Mereiter
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Álvaro M. Martins
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Catarina Gomes
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Meritxell Balmaña
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Joana A. Macedo
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Karol Polom
- Department of Surgical Oncology Medical University of Gdansk Poland
- General Surgery and Surgical Oncology Department University of Siena Italy
| | - Franco Roviello
- General Surgery and Surgical Oncology Department University of Siena Italy
| | - Ana Magalhães
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Celso A. Reis
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
- Faculty of Medicine University of Porto Portugal
- Instituto de Ciências Biomédicas Abel Salazar University of Porto Portugal
| |
Collapse
|
22
|
Sun Y, Yu X, Li M, Zou Z. Expression of CD44v6 and lymphatic vessel density in early gastric cancer tissues and their clinical significance. Pak J Med Sci 2019; 35:549-554. [PMID: 31086549 PMCID: PMC6500799 DOI: 10.12669/pjms.35.2.464] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Objective: To explore the relationships between expression of CD44v6, lymphatic vessel density (LVD) and the clinicopathological parameters of patients. Methods: One hundred early gastric cancer tissues, 55 high-grade gastric intraepithelial neoplasia (HGIN) tissues, 60 low-grade gastric intraepithelial neoplasia (LGIN) tissues and 60 chronic superficial gastritis tissues were collected and set as gastric cancer group, HGIN group, LGIN group and gastritis group respectively. The expression of CD44v6 and LVD of patients in all the groups were detected using two-step immunohistochemical method to analyze the relationships between the expression of CD44v6 and lymphatic vessel density in early gastric cancer tissues and their relationships with the clinicopathological parameters of patients. The values of LVD in predicting lymph node metastasis in early gastric cancer were evaluated using receiver operating characteristic (ROC) curve. Results: The positive expression of CD44v6 and LVD in the gastritis group, LGIN group, HGIN group and gastric cancer group gradually increased. The positive expression of CD44v6 and LVD in early gastric cancer tissues were in no correlation with the gender, age, tumor site, maximum diameter, differentiation degree and invasion depth (P>0.05) and in a correlation with lymphatic metastasis and lymphatic vessel invasion (P<0.06). The positive expression of CD44v6 in the early gastric cancer tissues was in a positive correlation with LVD (P<0.05). The analysis of ROC curves suggested that the area under ROC curve of predicting lymphatic metastasis of early gastric cancer with LVD was 0.837 (95% CI: 0.756~0.910), and the cut-off value was 14; the corresponding sensitivity and specificity were 63.6% and 90.2 respectively. Conclusion: The expression of CD44v6 and LVD in early gastric cancer tissues are in a close correlation with the clinicopathologic features, and joint detection of expression of CD44v6 and LVD can be taken as the indicator of gastric cancer metastasis.
Collapse
Affiliation(s)
- Yuting Sun
- Yuting Sun, Queen Mary College of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xiaowei Yu
- Xiaowei Yu, First Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Mengdi Li
- Mengdi Li, Queen Mary College of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Zhenhong Zou
- Zhenhong Zou, Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| |
Collapse
|
23
|
Kennedy PJ, Sousa F, Ferreira D, Pereira C, Nestor M, Oliveira C, Granja PL, Sarmento B. Fab-conjugated PLGA nanoparticles effectively target cancer cells expressing human CD44v6. Acta Biomater 2018; 81:208-218. [PMID: 30267881 DOI: 10.1016/j.actbio.2018.09.043] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 09/16/2018] [Accepted: 09/25/2018] [Indexed: 12/24/2022]
Abstract
Targeting of CD44 isoforms containing exon v6 (CD44v6) represents a viable strategy for the therapy and/or early diagnosis of metastatic cancers of the epithelium (e.g. gastric and colorectal cancer). We developed and characterized poly(lactic-co-glycolic acid) (PLGA)-based nanoparticles (NPs) modified with polyethylene glycol (PEG) and engrafted, by site-directed conjugation, with an engineered human Fab that specifically target human CD44v6 (v6 Fab-PLGA NPs). The v6 Fab-PLGA NPs displayed spherical morphology around 300 nm and were negatively charged. They strongly bound to a CD44v6-derived peptide and, more importantly, to cells that endogenously and exogenously express CD44v6, but not to non-expressing cells and cells expressing the standard isoform of CD44. The v6 Fab-PLGA NPs also recognized CD44v6 in tumor sections from cells grown subcutaneously within mice. The NPs had nominal cytotoxicity at 50 µg/mL and withstood simulated intestinal fluid exposure. Interestingly, v6 Fab-PLGA NPs cryopreserved in 10% trehalose and stored maintained specific cell binding. In conclusion, we envision NPs targeting CD44v6 as potential in vivo diagnostic agents and/or as anti-cancer agents in patients previously stratified with CD44v6+ carcinomas. STATEMENT OF SIGNIFICANCE: The v6 Fab-PLGA NPs displayed many favorable qualities as a potential CD44v6-targeted drug and/or diagnostic delivery agent. The NPs were designed for optimal ligand orientation and for immediate administration into humans. v6 Fab-PLGA NPs strongly bound to cells that endogenously and exogenously express CD44v6, but not to non-expressing cells and cells expressing the standard isoform of CD44. Binding ability was retained after freeze-drying and long-term storage, providing evidences on the stability of Fab-functionalized NPs. These NPs can potentially be used as an in vivo diagnostic from parenteral or oral/rectal administration.
Collapse
|
24
|
Amorim S, da Costa DS, Freitas D, Reis CA, Reis RL, Pashkuleva I, Pires RA. Molecular weight of surface immobilized hyaluronic acid influences CD44-mediated binding of gastric cancer cells. Sci Rep 2018; 8:16058. [PMID: 30375477 PMCID: PMC6207784 DOI: 10.1038/s41598-018-34445-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/30/2018] [Indexed: 01/02/2023] Open
Abstract
The physiological importance of the interactions between hyaluronic acid (HA) and its main membrane receptor, CD44, in pathological processes, e.g. cancer, is well recognized. However, these interactions are mainly studied in solution, whereas HA in the extracellular matrix (ECM) is partially immobilized via its interactions with other ECM components. We therefore, developed substrates in which HA is presented in an ECM-relevant manner. We immobilized HA with different molecular weights (Mw) in a Layer-by-Layer (LbL) fashion and studied the interactions of the substrates with CD44 and two human gastric cancer cell lines that overexpress this receptor, namely AGS and MKN45. We demonstrate that MKN45 cells are more sensitive to the LbL substrates as compared with AGS. This difference is due to different CD44 expression: while CD44 is detected mainly in the cytoplasm of AGS, MKN45 express CD44 predominantly at the cell membrane where it is involved in the recognition and binding of HA. The invasiveness of the studied cell lines was also evaluated as a function of HA Mw. Invasive profile characterized by low cell adhesion, high cell motility, high expression of cortactin, formation of invadopodia and cell clusters was observed for MKN45 cells when they are in contact with substrates presenting HA of high Mw.
Collapse
Affiliation(s)
- Sara Amorim
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| | - Diana Soares da Costa
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Daniela Freitas
- Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Celso A Reis
- Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
- Department of Pathology and Oncology, Faculty of Medicine, Porto University, Porto, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| | - Iva Pashkuleva
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Ricardo A Pires
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal.
| |
Collapse
|
25
|
Kennedy PJ, Perreira I, Ferreira D, Nestor M, Oliveira C, Granja PL, Sarmento B. Impact of surfactants on the target recognition of Fab-conjugated PLGA nanoparticles. Eur J Pharm Biopharm 2018; 127:366-370. [DOI: 10.1016/j.ejpb.2018.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 03/11/2018] [Accepted: 03/12/2018] [Indexed: 01/01/2023]
|
26
|
Lourenço BN, Springer NL, Ferreira D, Oliveira C, Granja PL, Fischbach C. CD44v6 increases gastric cancer malignant phenotype by modulating adipose stromal cell-mediated ECM remodeling. Integr Biol (Camb) 2018; 10:145-158. [PMID: 29450424 PMCID: PMC5988203 DOI: 10.1039/c7ib00179g] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
CD44, an abundantly expressed adhesion molecule, and its alternative splice variants have been associated with tumorigenesis and metastasis. In the context of gastric cancer (GC), de novo expression of CD44 variant 6 (CD44v6) is found in more than 60% of GCs, but its role in the pathogenesis and progression of this type of cancer remains unclear. Using a combination of media conditioning experiments and decellularized extracellular matrices (ECMs), this study investigates the hypothesis that CD44v6 overexpression enhances tumor cell malignant behavior by modulating stromal cell-mediated ECM remodeling. Our findings indicate that soluble factors secreted by CD44v6 expressing GC cells particularly increase proliferation and myofibroblastic differentiation of adipose stromal cells (ASCs). These changes in ASC phenotype mediate the deposition of fibrotic/desmoplastic ECM that, in turn, stimulates GC proliferation and inhibits GC clustering. Pharmacological inhibition of matrix metalloproteinase (MMP) activity in tumor cells abrogated matrix-induced changes in tumor cell malignant behavior. Additionally, studies in mice confirmed the pathological relevance of CD44v6 expression and consequential changes in ECM remodeling to gastric tumorigenesis in vivo. Collectively, these results indicate a direct link between CD44v6, ECM remodeling, and GC malignant behavior opening new insights into potential CD44v6-targeted therapies.
Collapse
Affiliation(s)
- Bianca N Lourenço
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, 157 Weill Hall, Ithaca, NY 14853, USA. and i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal and IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Portugal and Faculdade de Engenharia, Universidade do Porto, Portugal
| | - Nora L Springer
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, 157 Weill Hall, Ithaca, NY 14853, USA. and Biological and Biomedical Sciences, Cornell University, Ithaca, NY, USA
| | - Daniel Ferreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal and IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Portugal and Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Portugal
| | - Carla Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal and IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Portugal and Departamento de Patologia e Oncologia, Faculdade de Medicina, Universidade do Porto, Portugal
| | - Pedro L Granja
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal and INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Portugal and Faculdade de Engenharia, Universidade do Porto, Portugal and Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Portugal
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, 157 Weill Hall, Ithaca, NY 14853, USA. and Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY, USA
| |
Collapse
|
27
|
Zhang H, Feng M, Feng Y, Bu Z, Li Z, Jia S, Ji J. Germline mutations in hereditary diffuse gastric cancer. Chin J Cancer Res 2018; 30:122-130. [PMID: 29545726 DOI: 10.21147/j.issn.1000-9604.2018.01.13] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer is one of the leading causes of cancer-related deaths worldwide. Among which, about 1%-3% of gastric cancer patients were characterized by inherited gastric cancer predisposition syndromes, knowing as hereditary diffuse gastric cancer (HDGC). Studies reported that CDH1 germline mutations are the main cause of HDGC. With the help of rapid development of genetic testing technologies and data analysis tools, more and more researchers focus on seeking candidate susceptibility genes for hereditary cancer syndromes. In addition, National Comprehensive Cancer Network (NCCN) guidelines recommend that the patients of HDGC carrying CDH1 mutations should undergo prophylactic gastrectomy or routine endoscopic surveillances. Therefore, genetic counseling plays a key role in helping individuals with pathogenic mutations make appropriate risk management plans. Moreover, experienced and professional genetic counselors as well as a systematic multidisciplinary team (MDT) are also required to facilitate the development of genetic counseling and benefit pathogenic mutation carriers who are in need of regular and standardized risk management solutions. In this review, we provided an overview about the germline mutations of several genes identified in HDGC, suggesting that these genes may potentially act as susceptibility genes for this malignant cancer syndrome. Furthermore, we introduced information for prevention, diagnosis and risk management of HDGC. Investigations on key factors that may have effect on risk management decision-making and genetic data collection of more cancer syndrome family pedigrees are required for the development of HDGC therapeutic strategies.
Collapse
Affiliation(s)
- Hao Zhang
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Molecular Diagnostics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Mengmeng Feng
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Yi Feng
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Zhaode Bu
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Ziyu Li
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Shuqin Jia
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Molecular Diagnostics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jiafu Ji
- Surgery Laboratory, the Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| |
Collapse
|
28
|
|
29
|
Armero VES, Tremblay MP, Allaire A, Boudreault S, Martenon-Brodeur C, Duval C, Durand M, Lapointe E, Thibault P, Tremblay-Létourneau M, Perreault JP, Scott MS, Bisaillon M. Transcriptome-wide analysis of alternative RNA splicing events in Epstein-Barr virus-associated gastric carcinomas. PLoS One 2017; 12:e0176880. [PMID: 28493890 PMCID: PMC5426614 DOI: 10.1371/journal.pone.0176880] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/18/2017] [Indexed: 01/12/2023] Open
Abstract
Multiple human diseases including cancer have been associated with a dysregulation in RNA splicing patterns. In the current study, modifications to the global RNA splicing landscape of cellular genes were investigated in the context of Epstein-Barr virus-associated gastric cancer. Global alterations to the RNA splicing landscape of cellular genes was examined in a large-scale screen from 295 primary gastric adenocarcinomas using high-throughput RNA sequencing data. RT-PCR analysis, mass spectrometry, and co-immunoprecipitation studies were also used to experimentally validate and investigate the differential alternative splicing (AS) events that were observed through RNA-seq studies. Our study identifies alterations in the AS patterns of approximately 900 genes such as tumor suppressor genes, transcription factors, splicing factors, and kinases. These findings allowed the identification of unique gene signatures for which AS is misregulated in both Epstein-Barr virus-associated gastric cancer and EBV-negative gastric cancer. Moreover, we show that the expression of Epstein-Barr nuclear antigen 1 (EBNA1) leads to modifications in the AS profile of cellular genes and that the EBNA1 protein interacts with cellular splicing factors. These findings provide insights into the molecular differences between various types of gastric cancer and suggest a role for the EBNA1 protein in the dysregulation of cellular AS.
Collapse
Affiliation(s)
- Victoria E. S. Armero
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Marie-Pier Tremblay
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Andréa Allaire
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Simon Boudreault
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Camille Martenon-Brodeur
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Cyntia Duval
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Mathieu Durand
- Plateforme RNomique, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Elvy Lapointe
- Plateforme RNomique, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Philippe Thibault
- Plateforme RNomique, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Maude Tremblay-Létourneau
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Jean-Pierre Perreault
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Michelle S. Scott
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Martin Bisaillon
- Département de biochimie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
30
|
Demitrack ES, Samuelson LC. Notch as a Driver of Gastric Epithelial Cell Proliferation. Cell Mol Gastroenterol Hepatol 2017; 3:323-330. [PMID: 28462374 PMCID: PMC5404025 DOI: 10.1016/j.jcmgh.2017.01.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/28/2017] [Indexed: 02/08/2023]
Abstract
The gastric epithelium is sustained by a population of stem cells that replenish the various mature epithelial lineages throughout adulthood. Regulation of stem and progenitor cell proliferation occurs via basic developmental signaling pathways, including the Notch pathway, which recently was described to promote gastric stem cell proliferation in both mice and human beings. Current cancer theory proposes that adult stem cells that maintain gastrointestinal tissues accumulate mutations that promote cancerous growth, and that basic signaling pathways, such as Notch, which stimulate stem cell proliferation, can promote tumorigenesis. Accordingly, constitutive Notch activation leads to unchecked cellular proliferation and gastric tumors in genetic mouse models. Furthermore, there is emerging evidence suggesting that the Notch pathway may be activated in some human gastric cancers, supporting a potential role for Notch in gastric tumorigenesis. In this review, we first summarize the current understanding of gastric stem cells defined by genetic mouse studies, followed by discussion of the literature regarding Notch pathway regulation of gastric stem cell function in the mouse and human beings. Notch action to maintain gastric epithelial cell homeostasis and the cellular consequences of dysregulated signaling to promote tumorigenesis are discussed, including studies associating Notch activation with human gastric cancer. Finally, we compare and contrast Notch function in the stomach with other gastrointestinal tissues, including the intestine, to highlight the sensitivity of the stomach to Notch-induced tumors.
Collapse
Affiliation(s)
- Elise S. Demitrack
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Linda C. Samuelson
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
31
|
Zavros Y. Initiation and Maintenance of Gastric Cancer: A Focus on CD44 Variant Isoforms and Cancer Stem Cells. Cell Mol Gastroenterol Hepatol 2017; 4:55-63. [PMID: 28560289 PMCID: PMC5439237 DOI: 10.1016/j.jcmgh.2017.03.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/01/2017] [Indexed: 01/06/2023]
Abstract
Gastric cancer is the third most common cause of cancer-related death. Although the incidence of gastric cancer in the United States is relatively low, it remains significantly higher in some countries, including Japan and Korea. Interactions between cancer stem cells and the tumor microenvironment can have a substantial impact on tumor characteristics and contribute to heterogeneity. The mechanisms responsible for maintaining malignant cancer stem cells within the tumor microenvironment in human gastric cancer are largely unknown. Tumor cell and genetic heterogeneity contribute to either de novo intrinsic or the therapy-induced emergence of drug-resistant clones and eventual tumor recurrence. Although chemotherapy often is capable of inducing cell death in tumors, many cancer patients experience recurrence because of failure to effectively target the cancer stem cells, which are believed to be key tumor-initiating cells. Among the population of stem cells within the stomach that may be targeted during chronic Helicobacter pylori infection and altered into tumor-initiating cells are those cells marked by the cluster-of-differentiation (CD)44 cell surface receptor. CD44 variable isoforms (CD44v) have been implicated as key players in malignant transformation whereby their expression is highly restricted and specific, unlike the canonical CD44 standard isoform. Overall, CD44v, in particular CD44v9, are believed to mark the gastric cancer cells that contribute to increased resistance for chemotherapy- or radiation-induced cell death. This review focuses on the following: the alteration of the gastric stem cell during bacterial infection, and the role of CD44v in the initiation, maintenance, and growth of tumors associated with gastric cancer.
Collapse
Key Words
- CD, cluster-of-differentiation
- CD44v6
- CD44v9
- CD44v9, CD44 variant isoform containing exon v9
- CSC, cancer stem cell
- Cag, cytotoxin-associated gene
- Helicobacter pylori
- Inflammation
- Lgr5, leucine-rich, repeat-containing, G-protein–coupled receptor 5
- MDSC, myeloid-derived suppressor cell
- PDL1, programmed cell death 1 ligand
- PDTX, patient-derived tumor xenograft
- ROS, reactive oxygen species
- SPEM, spasmolytic polypeptide expressing metaplasia
- xCT, SLC7A11
Collapse
Affiliation(s)
- Yana Zavros
- Correspondence Address correspondence to: Yana Zavros, PhD, Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, 231 Albert B. Sabin Way, Room 4255 MSB, Cincinnati, Ohio 45267-0576. fax: (513) 558-5738.Department of Molecular and Cellular PhysiologyUniversity of Cincinnati College of Medicine231 Albert B. Sabin WayRoom 4255 MSBCincinnatiOhio 45267-0576
| |
Collapse
|
32
|
Nguyen PH, Giraud J, Chambonnier L, Dubus P, Wittkop L, Belleannée G, Collet D, Soubeyran I, Evrard S, Rousseau B, Senant-Dugot N, Mégraud F, Mazurier F, Varon C. Characterization of Biomarkers of Tumorigenic and Chemoresistant Cancer Stem Cells in Human Gastric Carcinoma. Clin Cancer Res 2016; 23:1586-1597. [PMID: 27620279 DOI: 10.1158/1078-0432.ccr-15-2157] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 07/19/2016] [Accepted: 07/31/2016] [Indexed: 12/14/2022]
Abstract
Purpose: Gastric carcinomas are heterogeneous, and the current therapy remains essentially based on surgery with conventional chemotherapy and radiotherapy. This study aimed to characterize biomarkers allowing the detection of cancer stem cells (CSC) in human gastric carcinoma of different histologic types.Experimental Design: The primary tumors from 37 patients with intestinal- or diffuse-type noncardia gastric carcinoma were studied, and patient-derived tumor xenograft (PDX) models in immunodeficient mice were developed. The expressions of 10 putative cell surface markers of CSCs, as well as aldehyde dehydrogenase (ALDH) activity, were studied, and the tumorigenic properties of cells were evaluated by in vitro tumorsphere assays and in vivo xenografts by limiting dilution assays.Results: We found that a subpopulation of gastric carcinoma cells expressing EPCAM, CD133, CD166, CD44, and a high ALDH activity presented the properties to generate new heterogeneous tumorspheres in vitro and tumors in vivo CD44 and CD166 were coexpressed, representing 6.1% to 37.5% of the cells; ALDH activity was detected in 1.6% to 15.4% of the cells; and the ALDH+ cells represented a core within the CD44+/CD166+ subpopulation that contained the highest frequency of tumorigenic CSCs in vivo The ALDH+ cells possessed drug efflux properties and were more resistant to standard chemotherapy than the ALDH- cells, a process that was partially reversed by verapamil treatment.Conclusions: CD44 and ALDH are the most specific biomarkers to detect and isolate tumorigenic and chemoresistant gastric CSCs in noncardia gastric carcinomas independently of the histologic classification of the tumor. Clin Cancer Res; 23(6); 1586-97. ©2016 AACR.
Collapse
Affiliation(s)
- Phu Hung Nguyen
- INSERM, U853 Helicobacter Infection, Inflammation and Cancer, Bordeaux, France.,University of Bordeaux, Bordeaux, France
| | - Julie Giraud
- INSERM, U853 Helicobacter Infection, Inflammation and Cancer, Bordeaux, France.,University of Bordeaux, Bordeaux, France
| | - Lucie Chambonnier
- INSERM, U853 Helicobacter Infection, Inflammation and Cancer, Bordeaux, France.,University of Bordeaux, Bordeaux, France
| | - Pierre Dubus
- University of Bordeaux, Bordeaux, France.,EA 2406, University of Bordeaux, Bordeaux, France.,University Hospital Center of Bordeaux, Bordeaux, France
| | - Linda Wittkop
- University of Bordeaux, Bordeaux, France.,INSERM, ISPED, Centre INSERM U1219 Bordeaux Population Health, Bordeaux, France.,Pôle de Santé Publique, Service d'information médicale, University Hospital Center of Bordeaux, Bordeaux, France
| | | | - Denis Collet
- University Hospital Center of Bordeaux, Bordeaux, France
| | - Isabelle Soubeyran
- Institut Bergonié, Bordeaux, France.,INSERM, U1012 Actions for onCogenesis understanding and Target Identification in Oncology (ACTION), Bordeaux, France
| | - Serge Evrard
- University of Bordeaux, Bordeaux, France.,Institut Bergonié, Bordeaux, France.,INSERM, U1012 Actions for onCogenesis understanding and Target Identification in Oncology (ACTION), Bordeaux, France
| | - Benoit Rousseau
- University of Bordeaux, Bordeaux, France.,Service Commun des Animaleries, Animalerie A2, Bordeaux, France
| | | | - Francis Mégraud
- INSERM, U853 Helicobacter Infection, Inflammation and Cancer, Bordeaux, France.,University of Bordeaux, Bordeaux, France.,University Hospital Center of Bordeaux, Bordeaux, France
| | | | - Christine Varon
- INSERM, U853 Helicobacter Infection, Inflammation and Cancer, Bordeaux, France. .,University of Bordeaux, Bordeaux, France
| |
Collapse
|
33
|
Branco da Cunha C, Klumpers DD, Koshy ST, Weaver JC, Chaudhuri O, Seruca R, Carneiro F, Granja PL, Mooney DJ. CD44 alternative splicing in gastric cancer cells is regulated by culture dimensionality and matrix stiffness. Biomaterials 2016; 98:152-62. [DOI: 10.1016/j.biomaterials.2016.04.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 04/15/2016] [Accepted: 04/18/2016] [Indexed: 12/19/2022]
|
34
|
Li Y, Yuan Y. Alternative RNA splicing and gastric cancer. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2016; 773:263-273. [PMID: 28927534 DOI: 10.1016/j.mrrev.2016.07.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 07/06/2016] [Accepted: 07/28/2016] [Indexed: 02/07/2023]
Abstract
Alternative splicing (AS) linked to diseases, especially to tumors. Recently, more and more studies focused on the relationship between AS and gastric cancer (GC). This review surveyed the hot topic from four aspects: First, the common types of AS in cancer, including exon skipping, intron retention, mutually exclusive exon, alternative 5 ' or 3' splice site, alternative first or last exon and alternative 3' untranslated regions. Second, basic mechanisms of AS and its relationship with cancer. RNA splicing in eukaryotes follows the GT-AG rule by both cis-elements and trans-acting factors regulatory. Through RNA splicing, different proteins with different forms and functions can be produced and may be associated with carcinogenesis. Third, AS types of GC-related genes and their splicing variants. In this paper, we listed 10 common genes with AS and illustrated its possible molecular mechanisms owing to genetic variation (mutation and /or polymorphism). Fourth, the splicing variants of GC-associated genes and gastric carcinogenesis, invasion and metastasis. Many studies have found that the different splicing variants of the same gene are differentially expressed in GC and its precancerous diseases, suggesting AS has important implications in GC development. Taking together, this review highlighted the role of AS and splicing variants in the process of GC. We hope that this is not only beneficial to advances in the study field of GC, but also can provide valuable information to other similar tumor research.Although we already know some gene splicing and splicing variants play an important role in the development of GC, but many phenomena and mechanisms are still unknown. For example, how the tumor microenvironment and signal transduction pathway effect the forming and function of AS? Unfortunately, this review did not cover the contents because the current study is limited. It is no doubt that clarifying the phenomena and mechanisms of these unknown may help to reveal the relationship of AS with complex tumor genetic variation and the occurrence and development of tumors.
Collapse
Affiliation(s)
- Ying Li
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Affiliated Hospital of China Medical University, Key Laboratory of Cancer Etiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang 110001, China
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Affiliated Hospital of China Medical University, Key Laboratory of Cancer Etiology and Prevention (China Medical University), Liaoning Provincial Education Department, Shenyang 110001, China.
| |
Collapse
|
35
|
Duarte HO, Freitas D, Gomes C, Gomes J, Magalhães A, Reis CA. Mucin-Type O-Glycosylation in Gastric Carcinogenesis. Biomolecules 2016; 6:E33. [PMID: 27409642 PMCID: PMC5039419 DOI: 10.3390/biom6030033] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 07/01/2016] [Accepted: 07/04/2016] [Indexed: 12/15/2022] Open
Abstract
Mucin-type O-glycosylation plays a crucial role in several physiological and pathological processes of the gastric tissue. Modifications in enzymes responsible for key glycosylation steps and the consequent abnormal biosynthesis and expression of their glycan products constitute well-established molecular hallmarks of disease state. This review addresses the major role played by mucins and associated O-glycan structures in Helicobacter pylori adhesion to the gastric mucosa and the subsequent establishment of a chronic infection, with concomitant drastic alterations of the gastric epithelium glycophenotype. Furthermore, alterations of mucin expression pattern and glycan signatures occurring in preneoplastic lesions and in gastric carcinoma are also described, as well as their impact throughout the gastric carcinogenesis cascade and in cancer progression. Altogether, mucin-type O-glycosylation alterations may represent promising biomarkers with potential screening and prognostic applications, as well as predictors of cancer patients' response to therapy.
Collapse
Affiliation(s)
- Henrique O Duarte
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Júlio Amaral de Carvalho, 45, Porto 4200-135, Portugal.
- Institute of Molecular Pathology and Immunology of University of Porto, Ipatimup, Rua Júlio Amaral de Carvalho, 45, Porto 4200-135, Portugal.
- Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo Ferreira no. 228, Porto 4050-313, Portugal.
| | - Daniela Freitas
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Júlio Amaral de Carvalho, 45, Porto 4200-135, Portugal.
- Institute of Molecular Pathology and Immunology of University of Porto, Ipatimup, Rua Júlio Amaral de Carvalho, 45, Porto 4200-135, Portugal.
- Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo Ferreira no. 228, Porto 4050-313, Portugal.
| | - Catarina Gomes
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Júlio Amaral de Carvalho, 45, Porto 4200-135, Portugal.
- Institute of Molecular Pathology and Immunology of University of Porto, Ipatimup, Rua Júlio Amaral de Carvalho, 45, Porto 4200-135, Portugal.
| | - Joana Gomes
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Júlio Amaral de Carvalho, 45, Porto 4200-135, Portugal.
- Institute of Molecular Pathology and Immunology of University of Porto, Ipatimup, Rua Júlio Amaral de Carvalho, 45, Porto 4200-135, Portugal.
| | - Ana Magalhães
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Júlio Amaral de Carvalho, 45, Porto 4200-135, Portugal.
- Institute of Molecular Pathology and Immunology of University of Porto, Ipatimup, Rua Júlio Amaral de Carvalho, 45, Porto 4200-135, Portugal.
| | - Celso A Reis
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Júlio Amaral de Carvalho, 45, Porto 4200-135, Portugal.
- Institute of Molecular Pathology and Immunology of University of Porto, Ipatimup, Rua Júlio Amaral de Carvalho, 45, Porto 4200-135, Portugal.
- Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), University of Porto, Rua Jorge Viterbo Ferreira no. 228, Porto 4050-313, Portugal.
- Medical Faculty, University of Porto, Alameda Prof Hernâni Monteiro, Porto 4200-319, Portugal.
| |
Collapse
|
36
|
Mereiter S, Balmaña M, Gomes J, Magalhães A, Reis CA. Glycomic Approaches for the Discovery of Targets in Gastrointestinal Cancer. Front Oncol 2016; 6:55. [PMID: 27014630 PMCID: PMC4783390 DOI: 10.3389/fonc.2016.00055] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 02/24/2016] [Indexed: 12/22/2022] Open
Abstract
Gastrointestinal (GI) cancer is the most common group of malignancies and many of its types are among the most deadly. Various glycoconjugates have been used in clinical practice as serum biomarker for several GI tumors, however, with limited diagnose application. Despite the good accessibility by endoscopy of many GI organs, the lack of reliable serum biomarkers often leads to late diagnosis of malignancy and consequently low 5-year survival rates. Recent advances in analytical techniques have provided novel glycoproteomic and glycomic data and generated functional information and putative biomarker targets in oncology. Glycosylation alterations have been demonstrated in a series of glycoconjugates (glycoproteins, proteoglycans, and glycosphingolipids) that are involved in cancer cell adhesion, signaling, invasion, and metastasis formation. In this review, we present an overview on the major glycosylation alterations in GI cancer and the current serological biomarkers used in the clinical oncology setting. We further describe recent glycomic studies in GI cancer, namely gastric, colorectal, and pancreatic cancer. Moreover, we discuss the role of glycosylation as a modulator of the function of several key players in cancer cell biology. Finally, we address several state-of-the-art techniques currently applied in this field, such as glycomic and glycoproteomic analyses, the application of glycoengineered cell line models, microarray and proximity ligation assay, and imaging mass spectrometry, and provide an outlook to future perspectives and clinical applications.
Collapse
Affiliation(s)
- Stefan Mereiter
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal; Institute of Biomedical Sciences of Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Meritxell Balmaña
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona , Girona , Spain
| | - Joana Gomes
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Ana Magalhães
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Celso A Reis
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal; Institute of Biomedical Sciences of Abel Salazar (ICBAS), University of Porto, Porto, Portugal; Medical Faculty, University of Porto, Porto, Portugal
| |
Collapse
|
37
|
van der Post RS, Gullo I, Oliveira C, Tang LH, Grabsch HI, O'Donovan M, Fitzgerald RC, van Krieken H, Carneiro F. Histopathological, Molecular, and Genetic Profile of Hereditary Diffuse Gastric Cancer: Current Knowledge and Challenges for the Future. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 908:371-91. [PMID: 27573781 DOI: 10.1007/978-3-319-41388-4_18] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Familial clustering is seen in 10 % of gastric cancer cases and approximately 1-3 % of gastric cancer arises in the setting of hereditary diffuse gastric cancer (HDGC). In families with HDGC, gastric cancer presents at young age. HDGC is predominantly caused by germline mutations in CDH1 and in a minority by mutations in other genes, including CTNNA1. Early stage HDGC is characterized by a few, up to dozens of intramucosal foci of signet ring cell carcinoma and its precursor lesions. These include in situ signet ring cell carcinoma and pagetoid spread of signet ring cells. Advanced HDGC presents as poorly cohesive/diffuse type carcinoma, normally with very few typical signet ring cells, and has a poor prognosis. Currently, it is unknown which factors drive the progression towards aggressive disease, but it is clear that most intramucosal lesions will not have such progression.Immunohistochemical profile of early and advanced HDGC is often characterized by abnormal E-cadherin immunoexpression, including absent or reduced membranous expression, as well as "dotted" or cytoplasmic expression. However, membranous expression of E-cadherin does not exclude HDGC. Intramucosal HDGC (pT1a) presents with an "indolent" phenotype, characterized by typical signet ring cells without immunoexpression of Ki-67 and p53, while advanced carcinomas (pT > 1) display an "aggressive" phenotype with pleomorphic cells, that are immunoreactive for Ki-67 and p53. These features show that the IHC profile is different between intramucosal and more advanced HDGC, providing evidence of phenotypic heterogeneity, and may help to define predictive biomarkers of progression from indolent to aggressive, widely invasive carcinomas.
Collapse
Affiliation(s)
- Rachel S van der Post
- Department of Pathology, Radboud University Medical Centre, 9101, Nijmegen, 6500 HB, The Netherlands
| | - Irene Gullo
- Department of Pathology, Centro Hospitalar de São João, Al. Prof. Hernâni Monteiro, Porto, 4200-319, Portugal.,Department of Pathology and Oncology, Faculdade de Medicina da Universidade do Porto (FMUP), Al. Prof. Hernâni Monteiro, Porto, 4200-319, Portugal.,Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Porto, Portugal and Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Dr. Roberto Frias S/N, Porto, 4200-465, Portugal
| | - Carla Oliveira
- Department of Pathology, Centro Hospitalar de São João, Al. Prof. Hernâni Monteiro, Porto, 4200-319, Portugal.,Department of Pathology and Oncology, Faculdade de Medicina da Universidade do Porto (FMUP), Al. Prof. Hernâni Monteiro, Porto, 4200-319, Portugal
| | - Laura H Tang
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave., New York, NY, 10065, USA
| | - Heike I Grabsch
- GROW School of Oncology and Developmental Biology and Department of Pathology, Maastricht University Medical Centre, Peter Debyelaan 25, Maastricht, 6229 HX, The Netherlands
| | - Maria O'Donovan
- Department of Histopathology, Cambridge University Hospitals NHS Trust, Cambridge, CB2 0QQ, UK
| | - Rebecca C Fitzgerald
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, 197, Biomedical Campus, Cambridge, CB2 0XZ, UK
| | - Han van Krieken
- Department of Pathology, Radboud University Medical Centre, 9101, Nijmegen, 6500 HB, The Netherlands
| | - Fátima Carneiro
- Department of Pathology, Centro Hospitalar de São João, Al. Prof. Hernâni Monteiro, Porto, 4200-319, Portugal. .,Department of Pathology and Oncology, Faculdade de Medicina da Universidade do Porto (FMUP), Al. Prof. Hernâni Monteiro, Porto, 4200-319, Portugal. .,Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Porto, Portugal and Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Dr. Roberto Frias S/N, Porto, 4200-465, Portugal.
| |
Collapse
|
38
|
CD44, Sonic Hedgehog, and Gli1 Expression Are Prognostic Biomarkers in Gastric Cancer Patients after Radical Resection. Gastroenterol Res Pract 2015; 2016:1013045. [PMID: 26839535 PMCID: PMC4709647 DOI: 10.1155/2016/1013045] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 10/25/2015] [Accepted: 11/16/2015] [Indexed: 02/07/2023] Open
Abstract
Aim. CD44 and Sonic Hedgehog (Shh) signaling are important for gastric cancer (GC). However, the clinical impact, survival, and recurrence outcome of CD44, Shh, and Gli1 expressions in GC patients following radical resection have not been elucidated. Patients and Methods. CD44, Shh, and Gli1 protein levels were quantified by immunohistochemistry (IHC). The association between CD44, Shh, and Gli1 expression and clinicopathological features or prognosis of GC patients was determined. The biomarker risk score was calculated by the IHC staining score of CD44, Shh, and Gli1 protein. Results. The IHC positive staining of CD44, Shh, and Gli1 proteins was correlated with larger tumour size, worse gross type and histological type, and advanced TNM stage, which also predicted shorter overall survival (OS) and disease-free survival (DFS) after radical resection. Multivariate analysis indicated the Gli1 protein and Gli1, CD44 proteins were predictive biomarkers for OS and DFS, respectively. If biomarker risk score was taken into analysis, it was the independent prognostic factor for OS and DFS. Conclusions. CD44 and Shh signaling are important biomarkers for tumour aggressiveness, survival, and recurrence in GC.
Collapse
|
39
|
Abstract
Despite recent progress in understanding the cancer genome, there is still a relative delay in understanding the full aspects of the glycome and glycoproteome of cancer. Glycobiology has been instrumental in relevant discoveries in various biological and medical fields, and has contributed to the deciphering of several human diseases. Glycans are involved in fundamental molecular and cell biology processes occurring in cancer, such as cell signalling and communication, tumour cell dissociation and invasion, cell-matrix interactions, tumour angiogenesis, immune modulation and metastasis formation. The roles of glycans in cancer have been highlighted by the fact that alterations in glycosylation regulate the development and progression of cancer, serving as important biomarkers and providing a set of specific targets for therapeutic intervention. This Review discusses the role of glycans in fundamental mechanisms controlling cancer development and progression, and their applications in oncology.
Collapse
Affiliation(s)
- Salomé S Pinho
- Instituto de Investigação e Inovação em Saúde (Institute for Research and Innovation in Health), University of Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira n.228, 4050-313 Porto, Portugal
| | - Celso A Reis
- Instituto de Investigação e Inovação em Saúde (Institute for Research and Innovation in Health), University of Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira n.228, 4050-313 Porto, Portugal
- Faculty of Medicine of the University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
40
|
Regulation of CD44E by DARPP-32-dependent activation of SRp20 splicing factor in gastric tumorigenesis. Oncogene 2015; 35:1847-56. [PMID: 26119931 PMCID: PMC4486340 DOI: 10.1038/onc.2015.250] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 05/11/2015] [Accepted: 05/22/2015] [Indexed: 12/19/2022]
Abstract
Objective CD44E is a frequently overexpressed variant of CD44 in gastric cancer. Mechanisms that regulate CD44 splicing and expression in gastric cancer remain unknown. Herein, we investigated the role of DARPP-32 (dopamine and cAMP-regulated phosphoprotein, Mr 32000) in promoting tumor growth through regulation of CD44 splicing. Design Quantitative luciferase reporter, quantitative real-time RT-PCR (qRT-PCR), Western blot, co-immunoprecipitation, ubiquitination, and tumor xenograft experiments were performed. Results Western blot and qRT-PCR results indicated that knockdown of endogenous DARPP-32 markedly reduces expression of CD44 V8-V10 (CD44E). Using a quantitative splicing luciferase reporter system, we detected a significant increase in the reporter activity following DARPP-32 overexpression (p < 0.001). Conversely, knocking down endogenous DARPP-32 significantly attenuated the splicing activity (p < 0.001). Further experiments showed that DARPP-32 regulates the expression of SRp20 splicing factor and co-exists with it in the same protein complex. Inhibition of alternative splicing with digitoxin followed by immunoprecipitation and immunoblotting indicated that DARPP-32 plays an important role in regulating SRp20 protein stability. The knockdown of endogenous DARPP-32 confirmed that DARPP-32 regulates the SRp20-dependent CD44E splicing. Using tumor xenograft mouse model, knocking down endogenous DARPP-32 markedly reduced SRp20 and CD44E protein levels with a decreased tumor growth. The reconstitution of SRp20 expression in these cells rescued tumor growth. In addition, we also demonstrated frequent co-overexpression and positive correlation of DARPP-32, SRp20 and CD44E expression levels in human gastric primary tumors. Conclusion Our novel findings establish for the first time the role of DARPP-32 in regulating splicing factors in gastric cancer cells. The DARPP-32–SRp20 axis plays a key role in regulating the CD44E splice variant that promotes gastric tumorigenesis.
Collapse
|
41
|
Campos D, Freitas D, Gomes J, Magalhães A, Steentoft C, Gomes C, Vester-Christensen MB, Ferreira JA, Afonso LP, Santos LL, Pinto de Sousa J, Mandel U, Clausen H, Vakhrushev SY, Reis CA. Probing the O-glycoproteome of gastric cancer cell lines for biomarker discovery. Mol Cell Proteomics 2015; 14:1616-29. [PMID: 25813380 PMCID: PMC4458724 DOI: 10.1074/mcp.m114.046862] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/19/2015] [Indexed: 12/21/2022] Open
Abstract
Circulating O-glycoproteins shed from cancer cells represent important serum biomarkers for diagnostic and prognostic purposes. We have recently shown that selective detection of cancer-associated aberrant glycoforms of circulating O-glycoprotein biomarkers can increase specificity of cancer biomarker assays. However, the current knowledge of secreted and circulating O-glycoproteins is limited. Here, we used the COSMC KO "SimpleCell" (SC) strategy to characterize the O-glycoproteome of two gastric cancer SimpleCell lines (AGS, MKN45) as well as a gastric cell line (KATO III) which naturally expresses at least partially truncated O-glycans. Overall, we identified 499 O-glycoproteins and 1236 O-glycosites in gastric cancer SimpleCells, and a total 47 O-glycoproteins and 73 O-glycosites in the KATO III cell line. We next modified the glycoproteomic strategy to apply it to pools of sera from gastric cancer and healthy individuals to identify circulating O-glycoproteins with the STn glycoform. We identified 37 O-glycoproteins in the pool of cancer sera, and only nine of these were also found in sera from healthy individuals. Two identified candidate O-glycoprotein biomarkers (CD44 and GalNAc-T5) circulating with the STn glycoform were further validated as being expressed in gastric cancer tissue. A proximity ligation assay was used to show that CD44 was expressed with the STn glycoform in gastric cancer tissues. The study provides a discovery strategy for aberrantly glycosylated O-glycoproteins and a set of O-glycoprotein candidates with biomarker potential in gastric cancer.
Collapse
Affiliation(s)
- Diana Campos
- From the ‡Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark; §IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| | - Daniela Freitas
- §IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| | - Joana Gomes
- §IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| | - Ana Magalhães
- §IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| | - Catharina Steentoft
- From the ‡Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Catarina Gomes
- §IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| | - Malene B Vester-Christensen
- From the ‡Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - José Alexandre Ferreira
- ¶Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Rua Dr. António Bernardino de Almeida 4200-072 Porto, Portugal; ‖QOPNA, Department of Chemistry of the University of Aveiro, Campus Universitário de Santiago 3810-193 Aveiro, Portugal
| | - Luis P Afonso
- **Department of Pathology, Portuguese Institute of Oncology, Rua Dr. António Bernardino de Almeida 4200-072 Porto, Portugal
| | - Lúcio L Santos
- ¶Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Rua Dr. António Bernardino de Almeida 4200-072 Porto, Portugal
| | - João Pinto de Sousa
- ‡‡Faculty of Medicine of the University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Ulla Mandel
- From the ‡Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Henrik Clausen
- From the ‡Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Sergey Y Vakhrushev
- From the ‡Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine and School of Dentistry, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark;
| | - Celso A Reis
- §IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal; ‡‡Faculty of Medicine of the University of Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; §§Institute of Biomedical Sciences Abel Salazar, ICBAS, Rua de Jorge Viterbo Ferreira n.228, 4050-313 Porto, Portugal
| |
Collapse
|
42
|
Bertaux-Skeirik N, Feng R, Schumacher MA, Li J, Mahe MM, Engevik AC, Javier JE, Peek Jr RM, Ottemann K, Orian-Rousseau V, Boivin GP, Helmrath MA, Zavros Y. CD44 plays a functional role in Helicobacter pylori-induced epithelial cell proliferation. PLoS Pathog 2015; 11:e1004663. [PMID: 25658601 PMCID: PMC4450086 DOI: 10.1371/journal.ppat.1004663] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 01/06/2015] [Indexed: 12/13/2022] Open
Abstract
The cytotoxin-associated gene (Cag) pathogenicity island is a strain-specific constituent of Helicobacter pylori (H. pylori) that augments cancer risk. CagA translocates into the cytoplasm where it stimulates cell signaling through the interaction with tyrosine kinase c-Met receptor, leading cellular proliferation. Identified as a potential gastric stem cell marker, cluster-of-differentiation (CD) CD44 also acts as a co-receptor for c-Met, but whether it plays a functional role in H. pylori-induced epithelial proliferation is unknown. We tested the hypothesis that CD44 plays a functional role in H. pylori-induced epithelial cell proliferation. To assay changes in gastric epithelial cell proliferation in relation to the direct interaction with H. pylori, human- and mouse-derived gastric organoids were infected with the G27 H. pylori strain or a mutant G27 strain bearing cagA deletion (∆CagA::cat). Epithelial proliferation was quantified by EdU immunostaining. Phosphorylation of c-Met was analyzed by immunoprecipitation followed by Western blot analysis for expression of CD44 and CagA. H. pylori infection of both mouse- and human-derived gastric organoids induced epithelial proliferation that correlated with c-Met phosphorylation. CagA and CD44 co-immunoprecipitated with phosphorylated c-Met. The formation of this complex did not occur in organoids infected with ∆CagA::cat. Epithelial proliferation in response to H. pylori infection was lost in infected organoids derived from CD44-deficient mouse stomachs. Human-derived fundic gastric organoids exhibited an induction in proliferation when infected with H. pylori that was not seen in organoids pre-treated with a peptide inhibitor specific to CD44. In the well-established Mongolian gerbil model of gastric cancer, animals treated with CD44 peptide inhibitor Pep1, resulted in the inhibition of H. pylori-induced proliferation and associated atrophic gastritis. The current study reports a unique approach to study H. pylori interaction with the human gastric epithelium. Here, we show that CD44 plays a functional role in H. pylori-induced epithelial cell proliferation.
Collapse
Affiliation(s)
- Nina Bertaux-Skeirik
- Department of Molecular and Cellular Physiology, University of Cincinnati,
Cincinnati, Ohio, United States of America
| | - Rui Feng
- Department of Molecular and Cellular Physiology, University of Cincinnati,
Cincinnati, Ohio, United States of America
| | - Michael A. Schumacher
- Department of Molecular and Cellular Physiology, University of Cincinnati,
Cincinnati, Ohio, United States of America
| | - Jing Li
- Department of Molecular and Cellular Physiology, University of Cincinnati,
Cincinnati, Ohio, United States of America
| | - Maxime M. Mahe
- Department of Surgery, Division of Pediatric Surgery, Cincinnati
Children’s Hospital Medical Center, Cincinnati, Ohio, United States of
America
| | - Amy C. Engevik
- Department of Molecular and Cellular Physiology, University of Cincinnati,
Cincinnati, Ohio, United States of America
| | - Jose E. Javier
- Department of Molecular and Cellular Physiology, University of Cincinnati,
Cincinnati, Ohio, United States of America
| | - Richard M. Peek Jr
- Cancer Biology, Vanderbilt University, Nashville, Tennessee, United States of
America
| | - Karen Ottemann
- Department of Microbiology and Environmental Toxicology, University of
California at Santa Cruz, Santa Cruz, California, United States of
America
| | - Veronique Orian-Rousseau
- Karlsruhe Institute of Technology, Institute for Toxicology and Genetics,
Hermann von Helmholtzplatz, Germany
| | - Gregory P. Boivin
- Department of Pathology Wright State University, Health Sciences, Dayton,
Ohio, United States of America
- Veterans Affairs Medical Center, Cincinnati, Ohio, United States of
America
| | - Michael A. Helmrath
- Department of Surgery, Division of Pediatric Surgery, Cincinnati
Children’s Hospital Medical Center, Cincinnati, Ohio, United States of
America
| | - Yana Zavros
- Department of Molecular and Cellular Physiology, University of Cincinnati,
Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
43
|
Zhao Y, Feng F, Zhou YN. Stem cells in gastric cancer. World J Gastroenterol 2015; 21:112-123. [PMID: 25574084 PMCID: PMC4284326 DOI: 10.3748/wjg.v21.i1.112] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 09/19/2014] [Accepted: 10/21/2014] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is one of the leading causes of cancer-related mortality worldwide. Cancer stem cells (CSCs), which were first identified in acute myeloid leukemia and subsequently in a large array of solid tumors, play important roles in cancer initiation, dissemination and recurrence. CSCs are often transformed tissue-specific stem cells or de-differentiated transit amplifying progenitor cells. Several populations of multipotent gastric stem cells (GSCs) that reside in the stomach have been determined to regulate physiological tissue renewal and injury repair. These populations include the Villin+ and Lgr5+ GSCs in the antrum, the Troy+ chief cells in the corpus, and the Sox2+ GSCs that are found in both the antrum and the corpus. The disruption of tumor suppressors in Villin+ or Lgr5+ GSCs leads to GC in mouse models. In addition to residing GSCs, bone marrow-derived cells can initiate GC in a mouse model of chronic Helicobacter infection. Furthermore, expression of the cell surface markers CD133 or CD44 defines gastric CSCs in mouse models and in human primary GC tissues and cell lines. Targeted elimination of CSCs effectively reduces tumor size and grade in mouse models. In summary, the recent identification of normal GSCs and gastric CSCs has greatly improved our understanding of the molecular and cellular etiology of GC and will aid in the development of effective therapies to treat patients.
Collapse
|
44
|
The ability of hyaluronan fragments to reverse the resistance of C6 rat glioma cell line to temozolomide and carmustine. Contemp Oncol (Pozn) 2014; 18:323-8. [PMID: 25477754 PMCID: PMC4248052 DOI: 10.5114/wo.2014.43493] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 12/11/2013] [Accepted: 04/11/2014] [Indexed: 11/18/2022] Open
Abstract
Aim of the study Hyaluronan (HA) is an extracellular matrix (ECM) polymer that may contribute to the emergence of anti-cancer drug resistance. Attempts to reverse drug resistance using small hyaluronan oligomers (oHA) are being made. The initial reports suggest that the oHA fraction may effectively reverse anti-cancer drug resistance in glioma models. However, the reversal effects of oHA of defined molecular length on glioma cells have not been investigated yet. In this study, we examined HA fragments containing 2 disaccharide units (oHA-2), 5 disaccharide units (oHA-5), and 68 kDa hyaluronan polymer (HA-68k) as agents possibly reversing the resistance of a C6 rat glioma cell line to temozolomide (TMZ) and carmustine (BCNU). Material and methods A 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) viability assay was used to assess the cytotoxicity of TMZ and BCNU in the presence or absence of the hyaluronan fragments. By comparing viability of the cells, the reversal effects of HA fragments on TMZ and BCNU resistance in C6 glioma cells were assessed. Results We found statistically significant decreases in the viability of cells in the presence of TMZ+oHA-5 as compared to TMZ alone (51.2 ±4.5 vs. 74.2 ±5.8, p = 0.0031), BCNU+o-HA5 as compared to BCNU alone (49.3 ±4.4 vs. 65.6 ±5.7, p = 0.0119), and BCNU+HA-68k as compared to BCNU alone (55.2 ±2.3 vs. 65.6 ±5.7, p = 0.0496). Conclusions Conclusions: Hyaluronan oligomers of 5 disaccharide units (oHA-5) significantly reversed the resistance of C6 cells to TMZ and BCNU. The results are only preliminary and a more thorough follow-up investigation is required to assess their actual role.
Collapse
|
45
|
Karbownik MS, Nowak JZ. Hyaluronan: towards novel anti-cancer therapeutics. Pharmacol Rep 2014; 65:1056-74. [PMID: 24399703 DOI: 10.1016/s1734-1140(13)71465-8] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 05/16/2013] [Indexed: 12/17/2022]
Abstract
The understanding of the role of hyaluronan in physiology and various pathological conditions has changed since the complex nature of its synthesis, degradation and interactions with diverse binding proteins was revealed. Initially perceived only as an inert component of connective tissue, it is now known to be involved in multiple signaling pathways, including those involved in cancer pathogenesis and progression. Hyaluronan presents a mixture of various length polymer molecules from finely fragmented oligosaccharides, polymers intermediate in size, to huge aggregates of high molecular weight hyaluronan. While large molecules promote tissue integrity and quiescence, the generation of breakdown products enhances signaling transduction, contributing to the pro-oncogenic behavior of cancer cells. Low molecular weight hyaluronan has well-established angiogenic properties, while the smallest hyaluronan oligomers may counteract tumor development. These equivocal properties make the role of hyaluronan in cancer biology very complex. This review surveys recent data on hyaluronan biosynthesis, metabolism, and interactions with its binding proteins called hyaladherins (CD44, RHAMM), providing themolecular background underlying its differentiated biological activity. In particular, the article critically presents current ideas on actual role of hyaluronan in cancer. The paper additionally maps a path towards promising novel anti-cancer therapeutics which target hyaluronan metabolic enzymes and hyaladherins, and constitute hyaluronan-based drug delivery systems.
Collapse
Affiliation(s)
- Michał S Karbownik
- Department of Pharmacology, Medical University of Lodz, Żeligowskiego 7/9, PL 90-752 Łódź, Poland. ;
| | | |
Collapse
|
46
|
Donnelly JM, Engevik A, Feng R, Xiao C, Boivin GP, Li J, Houghton J, Zavros Y. Mesenchymal stem cells induce epithelial proliferation within the inflamed stomach. Am J Physiol Gastrointest Liver Physiol 2014; 306:G1075-88. [PMID: 24789207 PMCID: PMC4059978 DOI: 10.1152/ajpgi.00489.2012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) sustain cancer cells by creating a microenvironment favorable for tumor growth. In particular, MSCs have been implicated in gastric cancer development. There is extensive evidence suggesting that Hedgehog signaling regulates tumor growth. However, very little is known regarding the precise roles of Hedgehog signaling and MSCs in tumor development within the stomach. The current study tests that hypothesis that Sonic Hedgehog (Shh), secreted from MSCs, provides a proliferative stimulus for the gastric epithelium in the presence of inflammation. Red fluorescent protein-expressing MSCs transformed in vitro (stMSCs) were transduced with lentiviral constructs containing a vector control (stMSC(vect)) or short hairpin RNA (shRNA) targeting the Shh gene (stMSC(ShhKO)). Gastric submucosal transplantation of wild-type MSCs (wtMSCs), wild-type MSCs overexpressing Shh (wtMSC(Shh)), stMSC(vect), or stMSC(ShhKO) cells in C57BL/6 control (BL/6) or gastrin-deficient (GKO) mice was performed and mice analyzed 30 and 60 days posttransplantation. Compared with BL/6 mice transplanted with wtMSC(Shh) and stMSC(vect) cells, inflamed GKO mice developed aggressive gastric tumors. Tumor development was not observed in mouse stomachs transplanted with wtMSC or stMSC(ShhKO) cells. Compared with stMSC(ShhKO)-transplanted mice, within the inflamed GKO mouse stomach, Shh-expressing stMSC(vect)- and wtMSC(Shh)-induced proliferation of CD44-positive cells. CD44-positive cells clustered in gland-like structures within the tumor stroma and were positive for Patched (Ptch) expression. We conclude that Shh, secreted from MSCs, provides a proliferative stimulus for the gastric epithelium that is associated with tumor development, a response that is sustained by chronic inflammation.
Collapse
Affiliation(s)
- Jessica M. Donnelly
- 1Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio;
| | - Amy Engevik
- 1Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio;
| | - Rui Feng
- 1Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio;
| | - Chang Xiao
- 2Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio;
| | - Gregory P. Boivin
- 3Department of Pathology Wright State University, Health Sciences, Dayton, Ohio; ,4Veterans Affairs Medical Center, Cincinnati, Ohio; and
| | - Jing Li
- 1Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio;
| | - JeanMarie Houghton
- 5Department of Medicine, Division of Gastroenterology, and Department of Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Yana Zavros
- Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio;
| |
Collapse
|
47
|
Sivadas VP, Gulati S, Varghese BT, Balan A, Kannan S. The early manifestation, tumor-specific occurrence and prognostic significance of TGFBR2 aberrant splicing in oral carcinoma. Exp Cell Res 2014; 327:156-62. [PMID: 24846200 DOI: 10.1016/j.yexcr.2014.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 05/07/2014] [Accepted: 05/10/2014] [Indexed: 12/22/2022]
Abstract
Alternative splicing is an important mechanism that can disrupt cell cycle control resulting in tumorigenesis. Although many alterations of Transforming Growth Factor Beta (TGFβ) signaling are reported in cancers, the role of splice aberrations in destabilizing this signaling is the least understood mechanism. In this study, we compared TGFBR2 alternative splicing events in potentially malignant oral disorders (PMDs) and oral squamous cell carcinoma (OSCC) samples with those in normal samples. Interestingly, there were five alternatively spliced forms of TGFBR2 with a deficient kinase domain in OSCCs. The TGFBR2 aberrant splicing was tumor-specific, suggesting that selective splicing out of TGFBR2 kinase domain could be a mechanism misused by cancer cells for evading TGFβ signaling-mediated anti-tumor activities. Moreover, these aberrant transcripts were present in PMDs as well, suggesting an early occurrence of these events during oral carcinogenesis and offering the possibility of early diagnosis of malignancy. Furthermore, OSCC patients who harbored these aberrantly spliced transcripts exhibited poor disease free survival (p=0.028) and poor overall survival (p=0.013). Thus, assessing the presence of these TGFBR2 transcripts can serve as a prognostic marker for oral cancer.
Collapse
Affiliation(s)
- V P Sivadas
- Division of Cancer Research, Regional Cancer Centre, Thiruvananthapuram 695011, Kerala, India
| | - Saakshi Gulati
- Department of Oral Medicine and Radiology, Govt. Dental College, Kozhikode, Kerala, India
| | - Bipin T Varghese
- Division of Surgical Oncology, Regional Cancer Centre, Thiruvananthapuram 695011, Kerala, India
| | - Anita Balan
- Department of Oral Medicine and Radiology, Govt. Dental College, Kozhikode, Kerala, India
| | - S Kannan
- Division of Cancer Research, Regional Cancer Centre, Thiruvananthapuram 695011, Kerala, India.
| |
Collapse
|
48
|
Guo SL, Ye H, Teng Y, Wang YL, Yang G, Li XB, Zhang C, Yang X, Yang ZZ, Yang X. Akt-p53-miR-365-cyclin D1/cdc25A axis contributes to gastric tumorigenesis induced by PTEN deficiency. Nat Commun 2014; 4:2544. [PMID: 24149576 PMCID: PMC3826643 DOI: 10.1038/ncomms3544] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 09/04/2013] [Indexed: 12/27/2022] Open
Abstract
Although PTEN/Akt signaling is frequently deregulated in human gastric cancers, the in vivo causal link between its dysregulation and gastric tumorigenesis has not been established. Here we show that inactivation of PTEN in mouse gastric epithelium initiates spontaneous carcinogenesis with complete penetrance by 2 months of age. Mechanistically, activation of Akt suppresses the abundance of p53, leading to decreased transcription of miR-365, thus causing upregulation of cyclin D1 and cdc25A, which promotes gastric cell proliferation. Importantly, genetic ablation of Akt1 restores miR-365 expression and effectively rescues gastric tumorigenesis in PTEN-mutant mice. Moreover, orthotopic restoration of miR-365 represses PTEN-deficient-induced hyperplasia. In human gastric cancer tissues, miR-365 reduction correlates with poorly differentiated histology, deep invasion and advanced stage, as well as the deregulation of PTEN, phosphorylated Akt, p53, cyclin D1 and cdc25A. These data demonstrate that the PTEN-Akt-p53-miR-365-cyclin D1/cdc25A axis serves as a new mechanism underlying gastric tumorigenesis, providing potential new therapeutic targets.
Collapse
Affiliation(s)
- Shui-Long Guo
- 1] State Key Laboratory of Proteomics, Genetic Laboratory of Development and Disease, Institute of Biotechnology, Beijing 100071, China [2] Institute of Geriatrics, PLA Postgraduate School of Medicine, PLA General Hospital, Beijing 100853, China [3]
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Lau WM, Teng E, Chong HS, Lopez KAP, Tay AYL, Salto-Tellez M, Shabbir A, So JBY, Chan SL. CD44v8-10 is a cancer-specific marker for gastric cancer stem cells. Cancer Res 2014; 74:2630-41. [PMID: 24618343 DOI: 10.1158/0008-5472.can-13-2309] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The surface marker CD44 has been identified as one of several markers associated with cancer stem cells (CSC) in solid tumors, but its ubiquitous expression in many cell types, including hematopoietic cells, has hindered its use in targeting CSCs. In this study, 28 paired primary tumor and adjacent nontumor gastric tissue samples were analyzed for cell surface protein expression. Cells that expressed pan-CD44 were found to occur at significantly higher frequency in gastric tumor tissues. We identified CD44v8-10 as the predominant CD44 variant expressed in gastric cancer cells and verified its role as a gastric CSC marker by limiting dilution and serial transplantation assays. Parallel experiments using CD133 failed to enrich for gastric CSCs. Analyses of another 26 primary samples showed significant CD44v8-10 upregulation in gastric tumor sites. Exogenous expression of CD44v8-10 but not CD44 standard (CD44s) increased the frequency of tumor initiation in immunocompromised mice. Reciprocal silencing of total CD44 resulted in reduced tumor-initiating potential of gastric cancer cells that could be rescued by CD44v8-10 but not CD44s expression. Our findings provide important functional evidence that CD44v8-10 marks human gastric CSCs and contributes to tumor initiation, possibly through enhancing oxidative stress defense. In addition, we showed that CD44v8-10 expression is low in normal tissues. Because CD44 also marks CSCs of numerous human cancers, many of which may also overexpress CD44v8-10, CD44v8-10 may provide an avenue to target CSCs in other human cancers.
Collapse
Affiliation(s)
- Wen Min Lau
- Authors' Affiliations: Cancer Science Institute of Singapore; Department of Surgery, National University of Singapore, Singapore, Singapore; and Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, Ireland, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Clinicopathologic significance of Sox2, CD44 and CD44v6 expression in intrahepatic cholangiocarcinoma. Pathol Oncol Res 2014; 20:655-60. [PMID: 24482053 DOI: 10.1007/s12253-014-9745-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 01/14/2014] [Indexed: 12/22/2022]
Abstract
Embryonic stem cells (ESC) and cancer stem cells (CSC) have a capacity for self-renewal and differentiation into multiple cell lineages. Sox2 plays a critical role in ESC and has been shown to participate in carcinogenesis and tumor progression in many human cancers. CD44 and CD44v6 are putative CSC markers and their association with tumor progression, metastasis, and tumor relapse after treatment has been demonstrated. We evaluated the immunoexpression of Sox2, CD44, and CD44v6 in 85 cases of Intrahepatic cholangiocarcinomas (IHCC) and assessed their prognostic significance. Sox2 expression showed a significant association with lymph node metastasis (p = 0.025), T4 stage (p = 0.046), and worse overall survival (p = 0.047). Greater expression of Sox2 was observed in IHCC with poor differentiation, vascular invasion, and stage IV, without statistical significance (p > 0.05). CD44 expression showed an association with periductal infiltrative type (p = 0.034), poor differentiation (p = 0.012), and vascular invasion (p = 0.009). CD44v6 expression was evident in patients with stage IV (p = 0.019). These results demonstrated that Sox2 expression is associated with aggressive behavior and poor overall survival in IHCC.
Collapse
|