1
|
Matoo S, Graves MJ, Choi MS, Idris RAES, Acharya P, Thapa G, Nguyen T, Atallah SY, Tipirneni AK, Stevenson PJ, Crawley SW. The microvillar protocadherin CDHR5 associates with EBP50 to promote brush border assembly. Mol Biol Cell 2024; 35:ar36. [PMID: 38170579 PMCID: PMC10916864 DOI: 10.1091/mbc.e23-02-0065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 10/27/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2024] Open
Abstract
Transporting epithelial cells of the gut and kidney interact with their luminal environment through a densely packed collection of apical microvilli known as a brush border (BB). Proper brush border assembly depends on the intermicrovillar adhesion complex (IMAC), a protocadherin-based adhesion complex found at the distal tips of microvilli that mediates adhesion between neighboring protrusions to promote their organized packing. Loss of the IMAC adhesion molecule Cadherin-related family member 5 (CDHR5) results in significant brush border defects, though the functional properties of this protocadherin have not been thoroughly explored. Here, we show that the cytoplasmic tail of CDHR5 contributes to its correct apical targeting and functional properties in an isoform-specific manner. Library screening identified the Ezrin-associated scaffolds EBP50 and E3KARP as cytoplasmic binding partners for CDHR5. Consistent with this, loss of EBP50 disrupted proper brush border assembly with cells exhibiting markedly reduced apical IMAC levels. Together, our results shed light on the apical targeting determinants of CDHR5 and further define the interactome of the IMAC involved in brush border assembly.
Collapse
Affiliation(s)
- Samaneh Matoo
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Maura J. Graves
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Myoung Soo Choi
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | | | - Prashun Acharya
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Garima Thapa
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Tram Nguyen
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Sarah Y. Atallah
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | - Ashna K. Tipirneni
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| | | | - Scott W. Crawley
- Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606
| |
Collapse
|
2
|
Słoka J, Madej M, Strzalka-Mrozik B. Molecular Mechanisms of the Antitumor Effects of Mesalazine and Its Preventive Potential in Colorectal Cancer. Molecules 2023; 28:5081. [PMID: 37446747 DOI: 10.3390/molecules28135081] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/18/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Chemoprevention is one of the ways to fight colorectal cancer, which is a huge challenge in oncology. Numerous pieces of evidence indicate that chronic inflammation in the course of Crohn's disease or ulcerative colitis (UC) is a significant cancer risk factor. Epidemiologic studies suggest that long-term use of non-steroidal anti-inflammatory drugs (NSAIDs), including mesalazine, has beneficial effects on colitis-associated colorectal cancer. Mesalazine is a first-line therapy for UC and is also widely used for maintaining remission in UC. Data showed that mesalazine has antiproliferative properties associated with cyclooxygenase (COX) inhibition but can also act through COX-independent pathways. This review summarizes knowledge about mesalazine's molecular mechanisms of action and chemopreventive effect by which it could interfere with colorectal cancer cell proliferation and survival.
Collapse
Affiliation(s)
- Joanna Słoka
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Marcel Madej
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Barbara Strzalka-Mrozik
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| |
Collapse
|
3
|
Bersuder E, Terciolo C, Lechevrel M, Martin E, Quesnelle C, Freund JN, Reimund JM, Gross I. Mesalazine initiates an anti-oncogenic β-catenin / MUCDHL negative feed-back loop in colon cancer cells by cell-specific mechanisms. Biomed Pharmacother 2021; 146:112543. [PMID: 34929577 DOI: 10.1016/j.biopha.2021.112543] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/06/2021] [Accepted: 12/13/2021] [Indexed: 01/18/2023] Open
Abstract
Chronic inflammation associated with intestinal architecture and barrier disruption puts patients with inflammatory bowel disease (IBD) at increased risk of developing colorectal cancer (CRC). Widely used to reduce flares of intestinal inflammation, 5-aminosalicylic acid derivatives (5-ASAs) such as mesalazine appear to also exert more direct mucosal healing and chemopreventive activities against CRC. The mechanisms underlying these activities are poorly understood and may involve the up-regulation of the cadherin-related gene MUCDHL (CDHR5). This atypical cadherin is emerging as a new actor of intestinal homeostasis and opposes colon tumorigenesis. Here, we showed that mesalazine increase mRNA levels of MUCDHL and of other genes involved in the intestinal barrier function in most intestinal cell lines. In addition, using gain / loss of function experiments (agonists, plasmid or siRNAs transfections), luciferase reporter genes and chromatin immunoprecipitation, we thoroughly investigated the molecular mechanisms triggered by mesalazine that lead to the up-regulation of MUCDHL expression. We found that basal transcription of MUCDHL in different CRC cell lines is regulated positively by CDX2 and negatively by β-catenin through a negative feed-back loop. However, mesalazine-stimulation of MUCDHL transcription is controlled by cell-specific mechanisms, involving either enhanced activation of CDX2 and PPAR-γ or repression of the β-catenin inhibitory effect. This work highlights the importance of the cellular and molecular context in the activity of mesalazine and suggests that its efficacy against CRC depends on the genetic alterations of transformed cells.
Collapse
Affiliation(s)
- Emilie Bersuder
- Université de Strasbourg, Inserm, IRFAC / UMR-S1113, FHU ARRIMAGE, FMTS, Strasbourg, France
| | - Chloe Terciolo
- Université de Strasbourg, Inserm, IRFAC / UMR-S1113, FHU ARRIMAGE, FMTS, Strasbourg, France
| | - Mathilde Lechevrel
- Université de Caen / Basse-Normandie, UFR de Médecine, EA 4652, F-14032 Caen, France
| | - Elisabeth Martin
- Université de Strasbourg, Inserm, IRFAC / UMR-S1113, FHU ARRIMAGE, FMTS, Strasbourg, France
| | - Celine Quesnelle
- Université de Caen / Basse-Normandie, UFR de Médecine, EA 4652, F-14032 Caen, France
| | - Jean-Noel Freund
- Université de Strasbourg, Inserm, IRFAC / UMR-S1113, FHU ARRIMAGE, FMTS, Strasbourg, France
| | - Jean-Marie Reimund
- Université de Strasbourg, Inserm, IRFAC / UMR-S1113, FHU ARRIMAGE, FMTS, Strasbourg, France; Université de Caen / Basse-Normandie, UFR de Médecine, EA 4652, F-14032 Caen, France; Service Hépato-Gastroentérologie, Hôpitaux Universitaires de Strasbourg, F-67000 Strasbourg, France; Institut Hospitalo-Universitaire de Strasbourg, Hôpitaux Universitaires de Strasbourg, F-67000 Strasbourg, France.
| | - Isabelle Gross
- Université de Strasbourg, Inserm, IRFAC / UMR-S1113, FHU ARRIMAGE, FMTS, Strasbourg, France.
| |
Collapse
|
4
|
Dutra TTB, Bezerra TMM, Luna ECM, Carvalho FSR, Chaves FN, Barros Silva PGD, Costa FWG, Pereira KMA. Do Protocadherins Show Prognostic Value in the Carcinogenesis of Human Malignant Neoplasms? Systematic Review and Meta-Analysis. Asian Pac J Cancer Prev 2020; 21:3677-3688. [PMID: 33369468 PMCID: PMC8046292 DOI: 10.31557/apjcp.2020.21.12.3677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Indexed: 12/24/2022] Open
Abstract
Background: Protocadherins (PCDHs) have been reported as tumor suppressor genes, implying that these genes may be involved in tumor suppression in a variety of cancers. However, a thorough understanding of the functions and mechanisms of PCDHs remains limited. Our aim was to investigate the methylation profile of PCDHs in human malignant neoplasms. Methods: This systematic review has been recorded in PROSPERO (#42019117844) and conducted according to PRISMA’s checklist; search was conducted in LILACS, PubMed, Science Direct, Scopus, and Web of Science databases, manually, with search queries and without date or language restrictions. Results: We found 91 articles, of which 26 were used for this meta-analysis and categorized according to the origin of the neoplasia. In total, 3,377 cases were compiled, with PCDH10, PCDH17, and PCDH8 being the most studied; males were 2.22 times more affected than females. Studies have shown significant heterogeneity (p <0.001), with the odds ratio varying between cases and controls [2.20 (95% CI = 1.11– 4.35) to 209.05 (95% CI = 12.64– 2,457.18)], and the value of association between methylation and cancers studied was 26.08 (95% CI = 15.42–44.13). Conclusion: In this systematic review, we have demonstrated using meta-analysis that PCDHs could emerge as potential tumor suppressor genes and that a significant increase in methylation may be useful for early detection of different cancers. This work may help in the identification of new prognostic biomarkers in malignant neoplasms.
Collapse
Affiliation(s)
- Thaís Torres Barros Dutra
- Department of Clinical Dentistry, Faculty of Pharmacy and Dentistry and Nursing, Federal University of Ceara, Fortaleza, Brazil
| | - Thâmara Manoela Marinho Bezerra
- Department of Clinical Dentistry, Faculty of Pharmacy and Dentistry and Nursing, Federal University of Ceara, Fortaleza, Brazil
| | - Ealber Carvalho Macêdo Luna
- Department of Clinical Dentistry, Faculty of Pharmacy and Dentistry and Nursing, Federal University of Ceara, Fortaleza, Brazil
| | | | - Filipe Nobre Chaves
- School of Dentistry, Federal University of Ceara, Campus Sobral, Sobral, Brazil
| | | | - Fábio Wildson Gurgel Costa
- Department of Clinical Dentistry, Faculty of Pharmacy and Dentistry and Nursing, Federal University of Ceara, Fortaleza, Brazil
| | | |
Collapse
|
5
|
Beck M, Baranger M, Moufok-Sadoun A, Bersuder E, Hinkel I, Mellitzer G, Martin E, Marisa L, Duluc I, de Reynies A, Gaiddon C, Freund JN, Gross I. The atypical cadherin MUCDHL antagonizes colon cancer formation and inhibits oncogenic signaling through multiple mechanisms. Oncogene 2020; 40:522-535. [PMID: 33188295 DOI: 10.1038/s41388-020-01546-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/12/2020] [Accepted: 10/28/2020] [Indexed: 01/24/2023]
Abstract
Cadherins form a large and pleiotropic superfamily of membranous proteins sharing Ca2+-binding repeats. While the importance of classic cadherins such as E- or N-cadherin for tumorigenesis is acknowledged, there is much less information about other cadherins that are merely considered as tissue-specific adhesion molecules. Here, we focused on the atypical cadherin MUCDHL that stood out for its unusual features and unique function in the gut. Analyses of transcriptomic data sets (n > 250) established that MUCDHL mRNA levels are down-regulated in colorectal tumors. Importantly, the decrease of MUCDHL expression is more pronounced in the worst-prognosis subset of tumors and is associated with decreased survival. Molecular characterization of the tumors indicated a negative correlation with proliferation-related processes (e.g., nucleic acid metabolism, DNA replication). Functional genomic studies showed that the loss of MUCDHL enhanced tumor incidence and burden in intestinal tumor-prone mice. Extensive structure/function analyses revealed that the mode of action of MUCDHL goes beyond membrane sequestration of ß-catenin and targets through its extracellular domain key oncogenic signaling pathways (e.g., EGFR, AKT). Beyond MUCDHL, this study illustrates how the loss of a gene critical for the morphological and functional features of mature cells contributes to tumorigenesis by dysregulating oncogenic pathways.
Collapse
Affiliation(s)
- Marine Beck
- Université de Strasbourg, Inserm, IRFAC UMR-S1113, 67200, Strasbourg, France
| | - Mathilde Baranger
- Université de Strasbourg, Inserm, IRFAC UMR-S1113, 67200, Strasbourg, France
| | - Ahlam Moufok-Sadoun
- Université de Strasbourg, Inserm, IRFAC UMR-S1113, 67200, Strasbourg, France
| | - Emilie Bersuder
- Université de Strasbourg, Inserm, IRFAC UMR-S1113, 67200, Strasbourg, France
| | - Isabelle Hinkel
- Université de Strasbourg, Inserm, IRFAC UMR-S1113, 67200, Strasbourg, France
| | - Georg Mellitzer
- Université de Strasbourg, Inserm, IRFAC UMR-S1113, 67200, Strasbourg, France
| | - Elisabeth Martin
- Université de Strasbourg, Inserm, IRFAC UMR-S1113, 67200, Strasbourg, France
| | | | - Isabelle Duluc
- Université de Strasbourg, Inserm, IRFAC UMR-S1113, 67200, Strasbourg, France
| | | | - Christian Gaiddon
- Université de Strasbourg, Inserm, IRFAC UMR-S1113, 67200, Strasbourg, France
| | - Jean-Noel Freund
- Université de Strasbourg, Inserm, IRFAC UMR-S1113, 67200, Strasbourg, France
| | - Isabelle Gross
- Université de Strasbourg, Inserm, IRFAC UMR-S1113, 67200, Strasbourg, France.
| |
Collapse
|
6
|
Gao J, Wang M, Li T, Liu Q, You L, Liao Q. Up-regulation of CDHR5 expression promotes malignant phenotype of pancreatic ductal adenocarcinoma. J Cell Mol Med 2020; 24:12726-12735. [PMID: 33025744 PMCID: PMC7687006 DOI: 10.1111/jcmm.15856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/30/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023] Open
Abstract
CDHR5 has been reported to play key roles in carcinogenesis of various cancers, but its roles in pancreatic cancer have not been reported. The present study was designed to investigate its clinical value in pancreatic ductal adenocarcinoma (PDAC). Tissue microarray-based immunohistochemistry was performed to analyse the correlation between CDHR5 expression and clinical and pathological features of PDAC, as well as the CDHR5 expression during tumour progression. Cell function assays were performed to investigate CDHR5's effects on PDAC cells. Moreover, qRT-PCR was applied to investigate the expression of CDHR5 isoforms in PDAC cells. Expression of CDHR5 was higher on the membrane of PDAC cells. This high expression level was associated with shorter overall survival of PDAC patients and was identified as an independent prognostic factor for overall survival by multivariate Cox regression analysis. In addition, expression level of CDHR5 presented an increased trend in the occurrence and progression of PDAC. Cell experiment suggested that CDHR5 could notably promote invasion and migration of PDAC cells. Moreover, analysis of CDHR5 isoforms indicated CDHR5-L was the major isoform expressed in PDAC cell lines. CDHR5 appears to be a promising and novel prognostic factor for PDAC, and its promotion in PDAC metastasis might be ascribed to the isoform CDHR5-L.
Collapse
Affiliation(s)
- Junyi Gao
- Department of General SurgeryPeking Union Medical College Hospital (PUMCH)Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC)BeijingChina
| | - Mengyi Wang
- Department of General SurgeryPeking Union Medical College Hospital (PUMCH)Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC)BeijingChina
| | - Tong Li
- Department of General SurgeryPeking Union Medical College Hospital (PUMCH)Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC)BeijingChina
| | - Qiaofei Liu
- Department of General SurgeryPeking Union Medical College Hospital (PUMCH)Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC)BeijingChina
| | - Lei You
- Department of General SurgeryPeking Union Medical College Hospital (PUMCH)Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC)BeijingChina
| | - Quan Liao
- Department of General SurgeryPeking Union Medical College Hospital (PUMCH)Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC)BeijingChina
| |
Collapse
|
7
|
Losi L, Zanocco-Marani T, Grande A. Cadherins down-regulation: towards a better understanding of their relevance in colorectal cancer. Histol Histopathol 2020; 35:1391-1402. [PMID: 32567668 DOI: 10.14670/hh-18-236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The down-regulation of cadherin expression in colorectal cancer (CRC) has been widely studied. However, existing data on cadherin expression are highly variable and its relevance to CRC development has not been completely established. This review examines published studies on cadherins whose down-regulation has been already demonstrated in CRC, trying to establish a relationship with promoter methylation, the capacity to influence the Wnt / CTNNB1 (catenin beta 1, beta-catenin) signalling pathway and the clinical implications for disease outcome. Moreover, it also analyses factors that may explain data variability and highlights the importance of considering the altered subcellular localization of the examined cadherins. The results of this survey reveal that thirty of one hundred existing cadherins appear to be down-regulated in CRC. Among these, ten are cadherins, sixteen are protocadherins, equally divided between clustered and non clustered, and four are cadherin - related. These findings suggest that, to better define the role played by cadherin down-regulation in CRC pathogenesis, the expression of multiple rather than individual cadherins should be taken into account and further functional studies are necessary to clarify the relative ability of individual cadherins to inhibit CTNNB1 therefore acting as tumor suppressors.
Collapse
Affiliation(s)
- Lorena Losi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| | | | - Alexis Grande
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
8
|
Ding X, Tian X, Liu W, Li Z. CDHR5 inhibits proliferation of hepatocellular carcinoma and predicts clinical prognosis. Ir J Med Sci 2019; 189:439-447. [PMID: 31482521 DOI: 10.1007/s11845-019-02092-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 08/27/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND As one of the most prevalent malignancies, hepatocellular carcinoma (HCC) ranks the third leading cause of cancer death worldwide. Due to the lack of biomarkers for early diagnosis, the clinical outcome of HCC remains unsatisfied with the current common therapeutic approaches, including surgery and chemotherapies. Thus, sensitive biomarkers and targeted therapies are in great need. AIMS In this study, we explored and verified whether CDHR5 (cadherin-related family member 5), a cadherin family protein, could serve as the potential biomarkers for HCC in the clinic. METHODS A retrospective study which contained 154 HCC patients was performed. Chi-square was utilized to analyze the relationship between CDHR5 expression and the clinicopathological features of HCC patients. The Kaplan-Meier method and Cox regression analyses were then used to evaluate the survival of HCC patients. In addition, cell proliferation assay and colony formation assay were performed to examine the effects of CDHR5 on the progression of HepG2 and Huh7 cells. RESULTS IHC and RT-qPCR revealed that CDHR5 was downregulated in HCC tissues compared with adjacent liver tissues. In addition, CDHR5 expression was significantly correlated with tumor numbers, tumor size, and TNM stage. CDHR5 expression was then shown to be an independent risk factor for survival of HCC patients by survival analysis. In vitro experiments showed that CDHR5 suppressed the proliferation capacity of HCC cells. CONCLUSIONS Taken together, our study not only identified CDHR5 as a novel prognostic biomarker in HCC but also provided evidence that CDHR5 can inhibit HCC cell proliferation.
Collapse
Affiliation(s)
- Xue Ding
- Department of Cardiology, Yidu Central Hospital of Weifang, Weifang, 262500, Shandong, China
| | - Xiaomin Tian
- Department of Cardiology, Yidu Central Hospital of Weifang, Weifang, 262500, Shandong, China
| | - Wei Liu
- Department of Cardiology, Yidu Central Hospital of Weifang, Weifang, 262500, Shandong, China
| | - Zijia Li
- Department of Immunology and Rheumatology, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
9
|
Loss of expression of μ-protocadherin and protocadherin-24 in sporadic and hereditary nonpolyposis colorectal cancers. Hum Pathol 2018; 84:299-308. [PMID: 30296522 DOI: 10.1016/j.humpath.2018.09.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/13/2018] [Accepted: 09/23/2018] [Indexed: 12/25/2022]
Abstract
Colorectal cancer (CRC) is a neoplastic disease in which normal mucosa undergoes a process of malignant transformation due to the progressive accumulation of molecular alterations affecting proto-oncogenes and oncosuppressor genes. Some of these modifications exert their carcinogenic potential by promoting a constitutive activation of the β-catenin signaling proliferation pathway, and when present, loss of cadherin expression also significantly contributes to the same effect. Using a combined approach of molecular and immunohistochemical analysis, we have previously demonstrated that most sporadic CRCs exhibit a down-regulated expression of a cadherin, named μ-protocadherin, that is generally observed in association with a higher proliferation rate and a worse prognosis. The aim of this report was to perform a comparative immunohistochemical assessment of μ-protocadherin and a similar cadherin, named protocadherin-24, in sporadic CRC and hereditary nonpolyposis colorectal cancer. The data obtained put in evidence that double-negative CRCs, lacking both the analyzed protocadherins, are more represented among sporadic tumors, whereas double-positive CRCs, maintaining their expression, exhibit an opposite trend. As expected, loss of protocadherin expression was accompanied by nuclear localization of β-catenin and increased positivity of the Ki-67 proliferation marker. This finding is consistent with the different clinical evolution of the 2 considered CRC sets according to which patients with hereditary nonpolyposis colorectal cancer experience a better prognosis as compared with those affected by a sporadic CRC.
Collapse
|
10
|
Piccoli M, D'Angelo E, Crotti S, Sensi F, Urbani L, Maghin E, Burns A, De Coppi P, Fassan M, Rugge M, Rizzolio F, Giordano A, Pilati P, Mammano E, Pucciarelli S, Agostini M. Decellularized colorectal cancer matrix as bioactive microenvironment for in vitro 3D cancer research. J Cell Physiol 2018; 233:5937-5948. [PMID: 29244195 DOI: 10.1002/jcp.26403] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 12/06/2017] [Indexed: 12/25/2022]
Abstract
Three-dimensional (3D) cancer models are overlooking the scientific landscape with the primary goal of bridging the gaps between two-dimensional (2D) cell lines, animal models and clinical research. Here, we describe an innovative tissue engineering approach applied to colorectal cancer (CRC) starting from decellularized human biopsies in order to generate an organotypic 3D-bioactive model. This in vitro 3D system recapitulates the ultrastructural environment of native tissue as demonstrated by histology, immunohistochemistry, immunofluorescence and scanning electron microscopy analyses. Mass spectrometry of proteome and secretome confirmed a different stromal composition between decellularized healthy mucosa and CRC in terms of structural and secreted proteins. Importantly, we proved that our 3D acellular matrices retained their biological properties: using CAM assay, we observed a decreased angiogenic potential in decellularized CRC compared with healthy tissue, caused by direct effect of DEFA3. We demonstrated that following a 5 days of recellularization with HT-29 cell line, the 3D tumor matrices induced an over-expression of IL-8, a DEFA3-mediated pathway and a mandatory chemokine in cancer growth and proliferation. Given the biological activity maintained by the scaffolds after decellularization, we believe this approach is a powerful tool for future pre-clinical research and screenings.
Collapse
Affiliation(s)
- Martina Piccoli
- Stem Cells and Regenerative Medicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Edoardo D'Angelo
- Nanoinspired Biomedicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
- First Surgical Clinic Section, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Sara Crotti
- Nanoinspired Biomedicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - Francesca Sensi
- Nanoinspired Biomedicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
- Department of Woman and Child Health, University of Padua, Padua, Italy
| | - Luca Urbani
- Stem Cells & Regenerative Medicine Section, Developmental Biology & Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
| | - Edoardo Maghin
- Stem Cells and Regenerative Medicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - Alan Burns
- Stem Cells & Regenerative Medicine Section, Developmental Biology & Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- Department of Clinical Genetics, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Paolo De Coppi
- Stem Cells & Regenerative Medicine Section, Developmental Biology & Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Padua, Italy
| | - Massimo Rugge
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Padua, Italy
- Veneto Tumor Registry, Padua, Italy
| | - Flavio Rizzolio
- Department of Translational Research, Pathology Unit, IRCCS-National Cancer Institute, Aviano, Italy
- Department of Molecular Sciences and Nanosystems at Ca' Foscari University, Venice, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | | | | | - Salvatore Pucciarelli
- First Surgical Clinic Section, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Marco Agostini
- Nanoinspired Biomedicine Lab, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
- First Surgical Clinic Section, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| |
Collapse
|
11
|
Parenti S, Montorsi L, Fantini S, Mammoli F, Gemelli C, Atene CG, Losi L, Frassineti C, Calabretta B, Tagliafico E, Ferrari S, Zanocco-Marani T, Grande A. KLF4 Mediates the Effect of 5-ASA on the β-Catenin Pathway in Colon Cancer Cells. Cancer Prev Res (Phila) 2018; 11:503-510. [PMID: 29794245 DOI: 10.1158/1940-6207.capr-17-0382] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 04/10/2018] [Accepted: 05/16/2018] [Indexed: 11/16/2022]
Abstract
Mesalazine (5-ASA) is an aminosalicylate anti-inflammatory drug capable of inducing μ-protocadherin, a protein expressed by colorectal epithelial cells that is downregulated upon malignant transformation. Treatment with 5-ASA restores μ-protocadherin expression and promotes the sequestration of β-catenin to the plasma membrane. Here, we show that 5-ASA-induced μ-protocadherin expression is directly regulated by the KLF4 transcription factor. In addition, we suggest the existence of a dual mechanism whereby 5-ASA-mediated β-catenin inhibition is caused by μ-protocadherin-dependent sequestration of β-catenin to the plasma membrane and by the direct binding of KLF4 to β-catenin. In addition, we found that 5-ASA treatment suppresses the expression of miR-130a and miR-135b, which target KLF4 mRNA, raising the possibility that this mechanism is involved in the increased expression of KLF4 induced by 5-ASA. Cancer Prev Res; 11(8); 503-10. ©2018 AACR.
Collapse
Affiliation(s)
- Sandra Parenti
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Lucia Montorsi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sebastian Fantini
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Fabiana Mammoli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Claudia Gemelli
- Science and Technology Park for Medicine, Mirandola, Modena, Italy
| | | | - Lorena Losi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Chiara Frassineti
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Bruno Calabretta
- Department of Clinical and Diagnostic Medicine and Public Health, University of Modena and Reggio Emilia, Modena, Italy.,Department of Cancer Biology and SKKC, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Enrico Tagliafico
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Sergio Ferrari
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Alexis Grande
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
12
|
Bläsius FM, Meller S, Stephan C, Jung K, Ellinger J, Glocker MO, Thiesen HJ, Tolkach Y, Kristiansen G. Loss of cadherin related family member 5 (CDHR5) expression in clear cell renal cell carcinoma is a prognostic marker of disease progression. Oncotarget 2017; 8:75076-75086. [PMID: 29088846 PMCID: PMC5650401 DOI: 10.18632/oncotarget.20507] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 07/29/2017] [Indexed: 11/25/2022] Open
Abstract
Reduced expression of Cadherin-Related Family Member 5 (CDHR5) was recently found implied in carcinogenesis of colon cancer, but its role in other tumors is unknown. We aimed to analyze the expression of CDHR5 in different subtypes of renal cell carcinoma. CDHR5 expression was immunohistochemically examined using tissue micro arrays (TMAs) covering 279 patients with primary renal cell carcinoma. Additionally, expression data from the TCGA (The Cancer Genome Atlas) of an independent cohort of 489 clear-cell RCC cases was evaluated. CDHR5 protein expression was found in 74.9% of cases, with higher levels seen in clear cell and papillary RCC. In the univariate analysis CDHR5 expression was significantly associated with a longer overall survival of RCC patients at the protein (p = 0.026, HR = 0.56) and transcript levels (TCGA-cohort: p = 0.0002, HR = 0.55). Importantly, differences in survival times were confirmed independently in multivariate analyses in a model with common clinicopathological variables at the transcript level (p = 0.0097, HR = 0.65). Investigation of the putative functional role of CDHR5 using TCGA data and Enrichment analysis for Gene Ontology and Pathways revealed associations with many metabolic and some tumor growth-associated processes and pathways. CDHR5 expression appears to be a promising and new independent prognostic biomarker in renal cell carcinoma.
Collapse
Affiliation(s)
| | | | - Carsten Stephan
- Clinic of Urology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Klaus Jung
- Berlin Institute for Urologic Research, Robert-Koch Platz 7, Berlin, Germany
| | - Jörg Ellinger
- Clinic of Urology, University Hospital Bonn, Bonn, Germany
| | | | | | - Yuri Tolkach
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | | |
Collapse
|
13
|
Montorsi L, Parenti S, Losi L, Ferrarini F, Gemelli C, Rossi A, Manco G, Ferrari S, Calabretta B, Tagliafico E, Zanocco-Marani T, Grande A. Expression of μ-protocadherin is negatively regulated by the activation of the β-catenin signaling pathway in normal and cancer colorectal enterocytes. Cell Death Dis 2016; 7:e2263. [PMID: 27310872 PMCID: PMC5143391 DOI: 10.1038/cddis.2016.163] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 05/05/2016] [Accepted: 05/05/2016] [Indexed: 01/11/2023]
Abstract
Mu-protocadherin (MUCDHL) is an adhesion molecule predominantly expressed by colorectal epithelial cells which is markedly downregulated upon malignant transformation. Notably, treatment of colorectal cancer (CRC) cells with mesalazine lead to increased expression of MUCDHL, and is associated with sequestration of β-catenin on the plasma membrane and inhibition of its transcriptional activity. To better characterize the causal relationship between β-catenin and MUCDHL expression, we performed various experiments in which CRC cell lines and normal colonic organoids were subjected to culture conditions inhibiting (FH535 treatment, transcription factor 7-like 2 siRNA inactivation, Wnt withdrawal) or stimulating (LiCl treatment) β-catenin activity. We show here that expression of MUCDHL is negatively regulated by functional activation of the β-catenin signaling pathway. This finding was observed in cell culture systems representing conditions of physiological stimulation and upon constitutive activation of β-catenin in CRC. The ability of MUCDHL to sequester and inhibit β-catenin appears to provide a positive feedback enforcing the effect of β-catenin inhibitors rather than serving as the primary mechanism responsible for β-catenin inhibition. Moreover, MUCDHL might have a role as biomarker in the development of CRC chemoprevention drugs endowed with β-catenin inhibitory activity.
Collapse
Affiliation(s)
- L Montorsi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - S Parenti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - L Losi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | - C Gemelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - A Rossi
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - G Manco
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - S Ferrari
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - B Calabretta
- Department of Clinical and Diagnostic Medicine and Public Health, University of Modena and Reggio Emilia, Modena, Italy.,Department of Cancer Biology and SKKC, Thomas Jefferson University, Philadelphia, PA, USA
| | - E Tagliafico
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - T Zanocco-Marani
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - A Grande
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
14
|
Woo YM, Shin Y, Hwang JA, Hwang YH, Lee S, Park EY, Kong HK, Park HC, Lee YS, Park JH. Epigenetic silencing of the MUPCDH gene as a possible prognostic biomarker for cyst growth in ADPKD. Sci Rep 2015; 5:15238. [PMID: 26463459 PMCID: PMC4604459 DOI: 10.1038/srep15238] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 08/28/2015] [Indexed: 11/09/2022] Open
Abstract
Although autosomal dominant polycystic kidney disease (ADPKD) is a common genetic disease, and is characterized by the formation of multiple fluid-filled cysts, which results in renal failure, early diagnosis and treatment of ADPKD have yet to be defined. Herein, we observed that the promoter region of the gene encoding mucin-like protocadherin (MUPCDH) was hypermethylated in the renal tissue of patients with ADPKD compared to non-ADPKD controls. Inversely, MUPCDH was significantly repressed in ADPKD, especially in cyst-lining cells. Our results indicate that aberrant methylation of MUPCDH promoter CpG islands may be negatively correlated with reduced expression level of MUPCDH and that this contributes to abnormal cell proliferation in ADPKD. It suggests that methylation status of MUPCDH promoter can be used as a novel epigenetic biomarker and a therapeutic target in ADPKD.
Collapse
Affiliation(s)
- Yu Mi Woo
- Department of Biological Science, Sookmyung Women's University, Seoul, 140-742, Korea
| | - Yubin Shin
- Department of Biological Science, Sookmyung Women's University, Seoul, 140-742, Korea
| | - Jung-Ah Hwang
- Branch of Cancer Genomics, Research Institute, National Cancer Center, Goyang-si, Gyeonggi-do, Korea
| | - Young-Hwan Hwang
- Department of Internal Medicine, Eulji General Hospital, Seoul, 139-892, Korea
| | - Sunyoung Lee
- Department of Biological Science, Sookmyung Women's University, Seoul, 140-742, Korea
| | - Eun Young Park
- Department of Biological Science, Sookmyung Women's University, Seoul, 140-742, Korea
| | - Hyun Kyung Kong
- Department of Biological Science, Sookmyung Women's University, Seoul, 140-742, Korea
| | - Hayne Cho Park
- Division of Nephrology, Armed Forces Capital Hospital, Seongnam, Korea
| | - Yeon-Su Lee
- Branch of Cancer Genomics, Research Institute, National Cancer Center, Goyang-si, Gyeonggi-do, Korea
| | - Jong Hoon Park
- Department of Biological Science, Sookmyung Women's University, Seoul, 140-742, Korea
| |
Collapse
|
15
|
Epigenetic-Mediated Downregulation of μ-Protocadherin in Colorectal Tumours. Gastroenterol Res Pract 2015; 2015:317093. [PMID: 25972897 PMCID: PMC4417986 DOI: 10.1155/2015/317093] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 03/25/2015] [Indexed: 11/18/2022] Open
Abstract
Carcinogenesis involves altered cellular interaction and tissue morphology that partly arise from aberrant expression of cadherins. Mucin-like protocadherin is implicated in intercellular adhesion and its expression was found decreased in colorectal cancer (CRC). This study has compared MUPCDH (CDHR5) expression in three key types of colorectal tissue samples, for normal mucosa, adenoma, and carcinoma. A gradual decrease of mRNA levels and protein expression was observed in progressive stages of colorectal carcinogenesis which are consistent with reports of increasing MUPCDH 5′ promoter region DNA methylation. High MUPCDH methylation was also observed in HCT116 and SW480 CRC cell lines that revealed low gene expression levels compared to COLO205 and HT29 cell lines which lack DNA methylation at the MUPCDH locus. Furthermore, HCT116 and SW480 showed lower levels of RNA polymerase II and histone H3 lysine 4 trimethylation (H3K4me3) as well as higher levels of H3K27 trimethylation at the MUPCDH promoter. MUPCDH expression was however restored in HCT116 and SW480 cells in the presence of 5-Aza-2′-deoxycytidine (DNA methyltransferase inhibitor). Results indicate that μ-protocadherin downregulation occurs during early stages of tumourigenesis and progression into the adenoma-carcinoma sequence. Epigenetic mechanisms are involved in this silencing.
Collapse
|
16
|
Bujko M, Kober P, Mikula M, Ligaj M, Ostrowski J, Siedlecki JA. Expression changes of cell-cell adhesion-related genes in colorectal tumors. Oncol Lett 2015; 9:2463-2470. [PMID: 26137091 DOI: 10.3892/ol.2015.3107] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 02/10/2015] [Indexed: 02/07/2023] Open
Abstract
Epithelial tissues achieve a highly organized structure due to cell-cell junction complexes. Carcinogenesis is accompanied by changes in cell interactions and tissue morphology, which appear in the early stages of benign tumors and progress along with invasive potential. The aim of the present study was to analyze the changes in expression levels of genes encoding intercellular junction proteins that have been previously identified to be differentially expressed in colorectal tumors compared with normal mucosa samples (fold change, >2.5) in genome-wide expression profiling. The expression of 20 selected genes was assessed using quantitative reverse transcription polymerase chain reaction in 26 colorectal cancer, 42 adenoma and 24 normal mucosa samples. Between these tissue types, differences were observed in the mRNA levels of genes encoding adherens junction proteins (upregulation of CDH3 and CDH11, and downregulation of CDH19 and PTPRF), tight junction proteins (upregulation of CLDN1 and CLDN2, and downregulation of CLDN5, CLDN8, CLDN23, CLDN15, JAM2 and CGN) and desmosomes (upregulation of DSC3 and DSG3, and downregulation of DSC2), in addition to a decrease in the expression of certain other genes involved in intercellular connections: PCDHB14, PCDH7, MUPCDH and NEO1. The differences between tissue types were statistically significant, and separate clustering of normal adenoma and carcinoma samples was observed in a hierarchical clustering analysis. These results indicate that the morphological changes in neoplastic colon tissue that occur during the 'adenoma-carcinoma sequence' are accompanied by specific changes in the expression of multiple genes encoding the majority of cell-cell junction complexes. The particular differential expression patterns appear to be consistent among patients with cancer and adenoma, in addition to normal mucosa samples.
Collapse
Affiliation(s)
- Mateusz Bujko
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw 02-781, Poland
| | - Paulina Kober
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw 02-781, Poland
| | - Michal Mikula
- Department of Genetics, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw 02-781, Poland
| | - Marcin Ligaj
- Department of Pathology, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw 02-781, Poland
| | - Jerzy Ostrowski
- Department of Genetics, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw 02-781, Poland
| | - Janusz Aleksander Siedlecki
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw 02-781, Poland
| |
Collapse
|
17
|
Hinkel I, Duluc I, Martin E, Guenot D, Freund JN, Gross I. Cdx2 controls expression of the protocadherin Mucdhl, an inhibitor of growth and β-catenin activity in colon cancer cells. Gastroenterology 2012; 142:875-885.e3. [PMID: 22202456 DOI: 10.1053/j.gastro.2011.12.037] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 11/28/2011] [Accepted: 12/09/2011] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS The intestine-specific homeobox transcription factor Cdx2 is an important determinant of intestinal identity in the embryonic endoderm and regulates the balance between proliferation and differentiation in the adult intestinal epithelium. Human colon tumors often lose Cdx2 expression, and heterozygous inactivation of Cdx2 in mice increases colon tumorigenesis. We sought to identify Cdx2 target genes to determine how it contributes to intestinal homeostasis. METHODS We used expression profiling analysis to identify genes that are regulated by Cdx2 in colon cancer cells lines. Regulation and function of a potential target gene were further investigated using various cell assays. RESULTS In colon cancer cell lines, Cdx2 directly regulated the transcription of the gene that encodes the protocadherin Mucdhl. Mucdhl localized to the apex of differentiated cells in the intestinal epithelium, and its expression was reduced in most human colon tumors. Overexpression of Mucdhl inhibited low-density proliferation of colon cancer cells and reduced tumor formation in nude mice. One isoform of Mucdhl interacted with β-catenin and inhibited its transcriptional activity. CONCLUSIONS The transcription factor Cdx2 activates expression of the protocadherin Mucdhl, which interacts with β-catenin and regulates activities of intestinal cells. Loss of Cdx2 expression in colon cancer cells might reduce expression of Mucdhl and thereby lead to tumor formation.
Collapse
|