Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 172
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell Commun Signal ; 22(1): 152, 2024 02 27.
Article in English | MEDLINE | ID: mdl-38414029

ABSTRACT

BACKGROUND: Germline mutations of E-cadherin contribute to hereditary diffuse gastric cancer (HDGC) and congenital malformations, such as oral facial clefts (OFC). However, the molecular mechanisms through which E-cadherin loss-of-function triggers distinct clinical outcomes remain unknown. We postulate that E-cadherin-mediated disorders result from abnormal interactions with the extracellular matrix and consequent aberrant intracellular signalling, affecting the coordination of cell migration. METHODS: Herein, we developed in vivo and in vitro models of E-cadherin mutants associated with either OFC or HDGC. Using a Drosophila approach, we addressed the impact of the different variants in cell morphology and migration ability. By combining gap closure migration assays and time-lapse microscopy, we further investigated the migration pattern of cells expressing OFC or HDGC variants. The adhesion profile of the variants was evaluated using high-throughput ECM arrays, whereas RNA sequencing technology was explored for identification of genes involved in aberrant cell motility. RESULTS: We have demonstrated that cells expressing OFC variants exhibit an excessive motility performance and irregular leading edges, which prevent the coordinated movement of the epithelial monolayer. Importantly, we found that OFC variants promote cell adhesion to a wider variety of extracellular matrices than HDGC variants, suggesting higher plasticity in response to different microenvironments. We unveiled a distinct transcriptomic profile in the OFC setting and pinpointed REG1A as a putative regulator of this outcome. Consistent with this, specific RNAi-mediated inhibition of REG1A shifted the migration pattern of OFC expressing cells, leading to slower wound closure with coordinated leading edges. CONCLUSIONS: We provide evidence that E-cadherin variants associated with OFC activate aberrant signalling pathways that support dynamic rearrangements of cells towards improved adaptability to the microenvironment. This proficiency results in abnormal tissue shaping and movement, possibly underlying the development of orofacial malformations.


Subject(s)
Adenocarcinoma , Stomach Neoplasms , Humans , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion , Cell Movement , Germ-Line Mutation , Lithostathine/genetics , Stomach Neoplasms/metabolism , Tumor Microenvironment , Animals , Drosophila melanogaster
2.
J Infect Dis ; 226(12): 2226-2237, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36173814

ABSTRACT

BACKGROUND: Helicobacter pylori infection induces cellular phenotypes relevant for cancer progression, namely cell motility and invasion. We hypothesized that the extracellular matrix (ECM) could be involved in these deleterious effects. METHODS: Microarrays were used to uncover ECM interactors in cells infected with H. pylori. LAMC2, encoding laminin γ2, was selected as a candidate gene and its expression was assessed in vitro and in vivo. The role of LAMC2 was investigated by small interference RNA (siRNA) combined with a set of functional assays. Laminin γ2 and E-cadherin expression patterns were evaluated in gastric cancer cases. RESULTS: Laminin γ2 was found significantly overexpressed in gastric cancer cells infected with H. pylori. This finding was validated in vitro by infection with clinical isolates and in vivo by using gastric biopsies of infected and noninfected individuals. We showed that laminin γ2 overexpression is dependent on the bacterial type IV secretion system and on the CagA. Functionally, laminin γ2 promotes cell invasion and resistance to apoptosis, through modulation of Src, JNK, and AKT activity. These effects were abrogated in cells with functional E-cadherin. CONCLUSIONS: These data highlight laminin γ2 and its downstream effectors as potential therapeutic targets, and the value of H. pylori eradication to delay gastric cancer onset and progression.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Humans , Helicobacter pylori/genetics , Laminin/metabolism , Helicobacter Infections/microbiology , Cell Line, Tumor , Cadherins/metabolism , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
3.
Gastric Cancer ; 25(1): 124-137, 2022 01.
Article in English | MEDLINE | ID: mdl-34486077

ABSTRACT

BACKGROUND: Tumour progression relies on the ability of cancer cells to penetrate and invade neighbouring tissues. E-cadherin loss is associated with increased cell invasion in gastric carcinoma, and germline mutations of the E-cadherin gene are causative of hereditary diffuse gastric cancer. Although E-cadherin dysfunction impacts cell-cell adhesion, cell dissemination also requires an imbalance of adhesion to the extracellular matrix (ECM). METHODS: To identify ECM components and receptors relevant for adhesion of E-cadherin dysfunctional cells, we implemented a novel ECM microarray platform coupled with molecular interaction networks. The functional role of putative candidates was determined by combining micropattern traction microscopy, protein modulation and in vivo approaches, as well as transcriptomic data of 262 gastric carcinoma samples, retrieved from the cancer genome atlas (TCGA). RESULTS: Here, we show that E-cadherin mutations induce an abnormal interplay of cells with specific components of the ECM, which encompasses increased traction forces and Integrin ß1 activation. Integrin ß1 synergizes with E-cadherin dysfunction, promoting cell scattering and invasion. The significance of the E-cadherin-Integrin ß1 crosstalk was validated in Drosophila models and found to be consistent with evidence from human gastric carcinomas, where increased tumour grade and poor survival are associated with low E-cadherin and high Integrin ß1 levels. CONCLUSIONS: Integrin ß1 is a key mediator of invasion in carcinomas with E-cadherin impairment and should be regarded as a biomarker of poor prognosis in gastric cancer.


Subject(s)
Integrin beta1 , Stomach Neoplasms , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion/physiology , Drosophila melanogaster , Extracellular Matrix/metabolism , Humans , Integrin beta1/genetics , Integrin beta1/metabolism , Neoplasm Invasiveness , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism
4.
Lancet Oncol ; 21(8): e386-e397, 2020 08.
Article in English | MEDLINE | ID: mdl-32758476

ABSTRACT

Hereditary diffuse gastric cancer (HDGC) is an autosomal dominant cancer syndrome that is characterised by a high prevalence of diffuse gastric cancer and lobular breast cancer. It is largely caused by inactivating germline mutations in the tumour suppressor gene CDH1, although pathogenic variants in CTNNA1 occur in a minority of families with HDGC. In this Policy Review, we present updated clinical practice guidelines for HDGC from the International Gastric Cancer Linkage Consortium (IGCLC), which recognise the emerging evidence of variability in gastric cancer risk between families with HDGC, the growing capability of endoscopic and histological surveillance in HDGC, and increased experience of managing long-term sequelae of total gastrectomy in young patients. To redress the balance between the accessibility, cost, and acceptance of genetic testing and the increased identification of pathogenic variant carriers, the HDGC genetic testing criteria have been relaxed, mainly through less restrictive age limits. Prophylactic total gastrectomy remains the recommended option for gastric cancer risk management in pathogenic CDH1 variant carriers. However, there is increasing confidence from the IGCLC that endoscopic surveillance in expert centres can be safely offered to patients who wish to postpone surgery, or to those whose risk of developing gastric cancer is not well defined.


Subject(s)
Neoplastic Syndromes, Hereditary , Stomach Neoplasms , Humans
5.
J Cell Mol Med ; 24(11): 5930-5936, 2020 06.
Article in English | MEDLINE | ID: mdl-32301282

ABSTRACT

E-cadherin protein (CDH1 gene) integrity is fundamental to the process of epithelial polarization and differentiation. Deregulation of the E-cadherin function plays a crucial role in breast cancer metastases, with worse prognosis and shorter overall survival. In this narrative review, we describe the inactivating mechanisms underlying CDH1 gene activity and its possible translation to clinical practice as a prognostic biomarker and as a potential targeted therapy.


Subject(s)
Breast Neoplasms/genetics , Cadherins/genetics , Gene Expression Regulation, Neoplastic , Biomarkers, Tumor/genetics , Cadherins/metabolism , Female , Humans , Molecular Targeted Therapy , Prognosis
6.
J Med Genet ; 56(4): 199-208, 2019 04.
Article in English | MEDLINE | ID: mdl-30661051

ABSTRACT

CDH1 encodes E-cadherin, a key protein in adherens junctions. Given that E-cadherin is involved in major cellular processes such as embryogenesis and maintenance of tissue architecture, it is no surprise that deleterious effects arise from its loss of function. E-cadherin is recognised as a tumour suppressor gene, and it is well established that CDH1 genetic alterations cause diffuse gastric cancer and lobular breast cancer-the foremost manifestations of the hereditary diffuse gastric cancer syndrome. However, in the last decade, evidence has emerged demonstrating that CDH1 mutations can be associated with lobular breast cancer and/or several congenital abnormalities, without any personal or family history of diffuse gastric cancer. To date, no genotype-phenotype correlations have been observed. Remarkably, there are reports of mutations affecting the same nucleotide but inducing distinct clinical outcomes. In this review, we bring together a comprehensive analysis of CDH1-associated disorders and germline alterations found in each trait, providing important insights into the biological mechanisms underlying E-cadherin's pleiotropic effects. Ultimately, this knowledge will impact genetic counselling and will be relevant to the assessment of risk of cancer development or congenital malformations in CDH1 mutation carriers.


Subject(s)
Antigens, CD/genetics , Cadherins/genetics , Cell Differentiation/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Germ-Line Mutation , Alleles , Breast Neoplasms/genetics , Cell Transformation, Neoplastic , Cleft Lip/genetics , Cleft Palate/genetics , Ectropion/genetics , Female , Genetic Association Studies/methods , Genotype , Humans , Male , Stomach Neoplasms/genetics , Tooth Abnormalities/genetics
7.
Cell Commun Signal ; 17(1): 155, 2019 11 25.
Article in English | MEDLINE | ID: mdl-31767037

ABSTRACT

BACKGROUND: E-cadherin has been awarded a key role in the aetiology of both sporadic and hereditary forms of gastric cancer. In this study, we aimed to identify molecular interactors that influence the expression and function of E-cadherin associated to cancer. METHODS: A data mining approach was used to predict stomach-specific candidate genes, uncovering S100P as a key candidate. The role of S100P was evaluated through in vitro functional assays and its expression was studied in a gastric cancer tissue microarray (TMA). RESULTS: S100P was found to contribute to a cancer pathway dependent on the context of E-cadherin function. In particular, we demonstrated that S100P acts as an E-cadherin positive regulator in a wild-type E-cadherin context, and its inhibition results in decreased E-cadherin expression and function. In contrast, S100P is likely to be a pro-survival factor in gastric cancer cells with loss of functional E-cadherin, contributing to an oncogenic molecular program. Moreover, expression analysis in a gastric cancer TMA revealed that S100P expression impacts negatively among patients bearing Ecad- tumours, despite not being significantly associated with overall survival on its own. CONCLUSIONS: We propose that S100P has a dual role in gastric cancer, acting as an oncogenic factor in the context of E-cadherin loss and as a tumour suppressor in a functional E-cadherin setting. The discovery of antagonist effects of S100P in different E-cadherin contexts will aid in the stratification of gastric cancer patients who may benefit from S100P-targeted therapies.


Subject(s)
Cadherins/metabolism , Calcium-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Stomach Neoplasms/metabolism , Cadherins/genetics , Calcium-Binding Proteins/genetics , Humans , Neoplasm Proteins/genetics , Signal Transduction/genetics , Stomach Neoplasms/pathology , Tumor Cells, Cultured
8.
J Med Genet ; 55(7): 431-441, 2018 07.
Article in English | MEDLINE | ID: mdl-29929997

ABSTRACT

Recent studies have reported germline CDH1 mutations in cases of lobular breast cancer (LBC) not associated with the classical hereditary diffuse gastric cancer syndrome. A multidisciplinary workgroup discussed genetic susceptibility, pathophysiology and clinical management of hereditary LBC (HLBC). The team has established the clinical criteria for CDH1 screening and results' interpretation, and created consensus guidelines regarding genetic counselling, breast surveillance and imaging techniques, clinicopathological findings, psychological and decisional support, as well as prophylactic surgery and plastic reconstruction. Based on a review of current evidence for the identification of HLBC cases/families, CDH1 genetic testing is recommended in patients fulfilling the following criteria: (A) bilateral LBC with or without family history of LBC, with age at onset <50 years, and (B) unilateral LBC with family history of LBC, with age at onset <45 years. In CDH1 asymptomatic mutant carriers, breast surveillance with clinical examination, yearly mammography, contrast-enhanced breast MRI and breast ultrasonography (US) with 6-month interval between the US and the MRI should be implemented as a first approach. In selected cases with personal history, family history of LBC and CDH1 mutations, prophylactic mastectomy could be discussed with an integrative group of clinical experts. Psychodecisional support also plays a pivotal role in the management of individuals with or without CDH1 germline alterations. Ultimately, the definition of a specific protocol for CDH1 genetic screening and ongoing coordinated management of patients with HLBC is crucial for the effective surveillance and early detection of LBC.


Subject(s)
Breast Neoplasms/genetics , Cadherins/genetics , Carcinoma, Lobular/genetics , Germ-Line Mutation/genetics , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Carcinoma, Lobular/diagnosis , Carcinoma, Lobular/pathology , Female , Genetic Counseling , Genetic Predisposition to Disease , Heterozygote , Humans , Mastectomy
9.
Mol Cancer ; 17(1): 112, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30068367

ABSTRACT

The aim of this study was to uncover the pathogenic relevance and the underlying molecular mechanism of a novel CDH1 variant found in a Hereditary Diffuse Gastric Cancer family (p.L13_L15del), which affects the signal peptide of E-cadherin without changing the remaining predicted sequence. We verified that p.L13_L15del cells yield low levels of E-cadherin, decreased cell adhesion and enhanced cell invasion. Further, we demonstrated that the disruption of the highly conserved hydrophobic core of the signal peptide hampers the binding of cellular components crucial for E-cadherin translation and translocation into the endoplasmic reticulum, constituting a new molecular basis for the loss of a tumour suppressor gene causative of hereditary cancer.


Subject(s)
Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Protein Sorting Signals , Stomach Neoplasms/genetics , Adult , Cell Adhesion , Endoplasmic Reticulum/metabolism , Female , Genetic Variation , Humans , Male , Protein Transport , Sequence Analysis, DNA , Stomach Neoplasms/metabolism
10.
Cell Commun Signal ; 16(1): 75, 2018 11 07.
Article in English | MEDLINE | ID: mdl-30404626

ABSTRACT

BACKGROUND: Basal-like breast cancer (BLBC) is a poor prognosis subgroup of triple-negative carcinomas that still lack specific target therapies and accurate biomarkers for treatment selection. P-cadherin is frequently overexpressed in these tumors, promoting cell invasion, stem cell activity and tumorigenesis by the activation of Src-Family kinase (SRC) signaling. Therefore, our aim was to evaluate if the treatment of BLBC cells with dasatinib, the FDA approved SRC inhibitor, would impact on P-cadherin induced tumor aggressive behavior. METHODS: P-cadherin and SRC expression was evaluated in a series of invasive Breast Cancer and contingency tables and chi-square tests were performed. Cell-cell adhesion measurements were performed by Atomic Force Microscopy, where frequency histograms and Gaussian curves were applied. 2D and 3D cell migration and invasion, proteases secretion and self-renew potential were evaluated in vitro. Student's t-tests were used to determine statistically significant differences. The cadherin/catenin complex interactions were evaluated by in situ proximity-ligation assay, and statistically significant results were determined by using Mann-Whitney test with a Bonferroni correction. In vivo xenograft mouse models were used to evaluate the impact of dasatinib on tumor growth and survival. ANOVA test was used to evaluate the differences in tumor size, considering a confidence interval of 95%. Survival curves were estimated by the Kaplan-Meier's method, using the log-rank test to assess significant differences for mice overall survival. RESULTS: Our data demonstrated that P-cadherin overexpression is significantly associated with SRC activation in breast cancer cells, which was also validated in a large series of primary tumor samples. SRC activity suppression with dasatinib significantly prevented the in vitro functional effects of P-cadherin overexpressing cells, as well as their in vivo tumorigenic and metastatic ability, by increasing mice overall survival. Mechanistically, SRC inhibition affects P-cadherin downstream signaling, rescues the E-cadherin/p120-catenin complex to the cell membrane, recovering cell-cell adhesion function. CONCLUSIONS: In conclusion our findings show that targeting P-cadherin/SRC signaling and functional activity may open novel therapeutic opportunities for highly aggressive and poor prognostic basal-like breast cancer.


Subject(s)
Breast Neoplasms/pathology , Cadherins/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , src-Family Kinases/antagonists & inhibitors , Animals , Carcinogenesis/drug effects , Catenins/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Dasatinib/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Neoplasm Metastasis , Delta Catenin
11.
Adv Exp Med Biol ; 1110: 35-53, 2018.
Article in English | MEDLINE | ID: mdl-30623365

ABSTRACT

Colorectal cancer (CRC) remains one of the leading causes of cancer mortality worldwide. Regarded as a heterogeneous disease, a number of biomarkers have been proposed to help in the stratification of CRC patients and to enable the selection of the best therapy for each patient towards personalized therapy. However, although the molecular mechanisms underlying the development of CRC have been elucidated, the therapeutic strategies available for these patients are still quite limited. Thus, over the last few years, a multitude of novel targets and therapeutic strategies have emerged focusing on deregulated molecules and pathways that are implicated in cell growth and survival. Particularly relevant in CRC are the activating mutations in the oncogene PIK3CA that frequently occur in concomitancy with KRAS and BRAF mutations and that lead to deregulation of the major signalling pathways PI3K and MAPK, downstream of EGFR. This review focus on the importance of the PI3K signalling in CRC development, on the current knowledge of PI3K inhibition as a therapeutic approach in CRC and on the implications PI3K signalling molecules may have as potential biomarkers and as new targets for directed therapies in CRC patients.


Subject(s)
Colorectal Neoplasms/drug therapy , Molecular Targeted Therapy , Phosphatidylinositol 3-Kinases , Signal Transduction , Humans , Mutation , Proto-Oncogene Proteins B-raf , Proto-Oncogene Proteins p21(ras)
12.
Lab Invest ; 97(5): 615-625, 2017 05.
Article in English | MEDLINE | ID: mdl-28263290

ABSTRACT

In the past decades, there has been an amazing progress in the understanding of the molecular mechanisms of the cell cycle. This has been possible largely due to a better conceptualization of the cycle itself, but also as a consequence of technological advances. Herein, we propose a new fluorescence image-based framework targeted at the identification and segmentation of stained nuclei with the purpose to determine DNA content in distinct cell cycle stages. The method is based on discriminative features, such as total intensity and area, retrieved from in situ stained nuclei by fluorescence microscopy, allowing the determination of the cell cycle phase of both single and sub-population of cells. The analysis framework was built on a modified k-means clustering strategy and refined with a Gaussian mixture model classifier, which enabled the definition of highly accurate classification clusters corresponding to G1, S and G2 phases. Using the information retrieved from area and fluorescence total intensity, the modified k-means (k=3) cluster imaging framework classified 64.7% of the imaged nuclei, as being at G1 phase, 12.0% at G2 phase and 23.2% at S phase. Performance of the imaging framework was ascertained with normal murine mammary gland cells constitutively expressing the Fucci2 technology, exhibiting an overall sensitivity of 94.0%. Further, the results indicate that the imaging framework has a robust capacity to both identify a given DAPI-stained nucleus to its correct cell cycle phase, as well as to determine, with very high probability, true negatives. Importantly, this novel imaging approach is a non-disruptive method that allows an integrative and simultaneous quantitative analysis of molecular and morphological parameters, thus awarding the possibility of cell cycle profiling in cytological and histological samples.


Subject(s)
Cell Cycle/physiology , Fluorescent Dyes/analysis , Indoles/analysis , Microscopy, Fluorescence/methods , Single-Cell Analysis/methods , Animals , Cell Line , Cell Nucleus/chemistry , Fluorescent Dyes/chemistry , Histocytochemistry , Image Processing, Computer-Assisted , Indoles/chemistry , Mice , Sensitivity and Specificity
13.
Hum Mol Genet ; 24(20): 5891-900, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26246502

ABSTRACT

Epithelial-cadherin (Ecad) deregulation affects cell-cell adhesion and results in increased invasiveness of distinct human carcinomas. In gastric cancer, loss of Ecad expression is a common event and is associated with disease aggressiveness and poor prognosis. However, the molecular mechanisms underlying the invasive process associated to Ecad dysfunction are far from understood. We hypothesized that deregulation of cell-matrix interactions could play an important role during this process. Thus, we focussed on LM-332, which is a major matrix component, and in Ecad/LM-332 crosstalk in the process of Ecad-dependent invasion. To verify whether matrix deregulation was triggered by Ecad loss, we used the Drosophila model. To dissect the key molecules involved and unveil their functional significance, we used gastric cancer cell lines. The relevance of this relationship was then confirmed in human primary tumours. In vivo, Ecad knockdown induced apoptosis; nonetheless, at the invasive front, cells ectopically expressed Laminin A and ßPS integrin. In vitro, we demonstrated that, in two different gastric cancer cell models, Ecad-defective cells overexpressed Laminin γ2 (LM-γ2), ß1 and ß4 integrin, when compared with Ecad-competent ones. We showed that LM-γ2 silencing impaired invasion and enhanced cell death, most likely via pSrc and pAkt reduction, and JNK activation. In human gastric carcinomas, we found a concomitant decrease in Ecad and increase in LM-γ2. This is the first evidence that ectopic Laminin expression depends on Ecad loss and allows Ecad-dysfunctional cells to survive and invade. This opens new avenues for using LM-γ2 signalling regulators as molecular targets to impair gastric cancer progression.


Subject(s)
Cadherins/genetics , Gene Deletion , Laminin/genetics , Stomach Neoplasms/metabolism , Animals , Cell Line, Tumor , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Humans , Neoplasm Invasiveness , Stomach Neoplasms/pathology , Stomach Neoplasms/physiopathology , Up-Regulation
14.
Int J Mol Sci ; 18(12)2017 Dec 12.
Article in English | MEDLINE | ID: mdl-29231860

ABSTRACT

The role of E-cadherin in Hereditary Diffuse Gastric Cancer (HDGC) is unequivocal. Germline alterations in its encoding gene (CDH1) are causative of HDGC and occur in about 40% of patients. Importantly, while in most cases CDH1 alterations result in the complete loss of E-cadherin associated with a well-established clinical impact, in about 20% of cases the mutations are of the missense type. The latter are of particular concern in terms of genetic counselling and clinical management, as the effect of the sequence variants in E-cadherin function is not predictable. If a deleterious variant is identified, prophylactic surgery could be recommended. Therefore, over the last few years, intensive research has focused on evaluating the functional consequences of CDH1 missense variants and in assessing E-cadherin pathogenicity. In that context, our group has contributed to better characterize CDH1 germline missense variants and is now considered a worldwide reference centre. In this review, we highlight the state of the art methodologies to categorize CDH1 variants, as neutral or deleterious. This information is subsequently integrated with clinical data for genetic counseling and management of CDH1 variant carriers.


Subject(s)
Cadherins/genetics , Genetic Predisposition to Disease/genetics , Mutation, Missense , Stomach Neoplasms/genetics , Antigens, CD , Cell Adhesion/genetics , Cell Movement/genetics , Genetic Counseling , Germ-Line Mutation , Heterozygote , Humans , Stomach Neoplasms/pathology
15.
Hum Mol Genet ; 23(8): 2094-105, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24293545

ABSTRACT

E-cadherin (Ecad) is a well-known invasion suppressor and its loss of expression is common in invasive carcinomas. Germline Ecad mutations are the only known genetic cause of hereditary diffuse gastric cancer (HDGC), demonstrating the causative role of Ecad impairment in gastric cancer. HDGC-associated Ecad missense mutations can lead to folding defects and premature proteasome-dependent endoplasmic reticulum-associated degradation (ERAD), but the molecular determinants for this fate were unidentified. Using a Drosophila-based genetic screen, we found that Drosophila DnaJ-1 interacts with wild type (WT) and mutant human Ecad in vivo. DnaJ (Hsp40) homolog, subfamily B, member 4 (DNAJB4), the human homolog of DnaJ-1, influences Ecad localization and stability even in the absence of Ecad endogenous promoter, suggesting a post-transcriptional level of regulation. Increased expression of DNAJB4 leads to stabilization of WT Ecad in the plasma membrane, while it induces premature degradation of unfolded HDGC mutants in the proteasome. The interaction between DNAJB4 and Ecad is direct, and is increased in the context of the unfolded mutant E757K, especially when proteasome degradation is inhibited, suggesting that DNAJB4 is a molecular mediator of ERAD. Post-translational regulation of native Ecad by DNAJB4 molecular chaperone is sufficient to influence cell adhesion in vitro. Using a chick embryo chorioallantoic membrane assay with gastric cancer derived cells, we demonstrate that DNAJB4 stimulates the anti-invasive function of WT Ecad in vivo. Additionally, the expression of DNAJB4 and Ecad is concomitantly decreased in human gastric carcinomas. Altogether, we demonstrate that DNAJB4 is a sensor of Ecad structural features that might contribute to gastric cancer progression.


Subject(s)
Animals, Genetically Modified/metabolism , Cadherins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , HSP40 Heat-Shock Proteins/metabolism , Mutation/genetics , Stomach Neoplasms/metabolism , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/growth & development , Blotting, Western , Cadherins/metabolism , Cell Membrane/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Chick Embryo , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Flow Cytometry , HSP40 Heat-Shock Proteins/genetics , Humans , Immunoenzyme Techniques , Immunoprecipitation , In Vitro Techniques , Molecular Chaperones/metabolism , Neoplasm Invasiveness , Proteolysis , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
16.
J Med Genet ; 52(6): 361-74, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25979631

ABSTRACT

Germline CDH1 mutations confer a high lifetime risk of developing diffuse gastric (DGC) and lobular breast cancer (LBC). A multidisciplinary workshop was organised to discuss genetic testing, surgery, surveillance strategies, pathology reporting and the patient's perspective on multiple aspects, including diet post gastrectomy. The updated guidelines include revised CDH1 testing criteria (taking into account first-degree and second-degree relatives): (1) families with two or more patients with gastric cancer at any age, one confirmed DGC; (2) individuals with DGC before the age of 40 and (3) families with diagnoses of both DGC and LBC (one diagnosis before the age of 50). Additionally, CDH1 testing could be considered in patients with bilateral or familial LBC before the age of 50, patients with DGC and cleft lip/palate, and those with precursor lesions for signet ring cell carcinoma. Given the high mortality associated with invasive disease, prophylactic total gastrectomy at a centre of expertise is advised for individuals with pathogenic CDH1 mutations. Breast cancer surveillance with annual breast MRI starting at age 30 for women with a CDH1 mutation is recommended. Standardised endoscopic surveillance in experienced centres is recommended for those opting not to have gastrectomy at the current time, those with CDH1 variants of uncertain significance and those that fulfil hereditary DGC criteria without germline CDH1 mutations. Expert histopathological confirmation of (early) signet ring cell carcinoma is recommended. The impact of gastrectomy and mastectomy should not be underestimated; these can have severe consequences on a psychological, physiological and metabolic level. Nutritional problems should be carefully monitored.


Subject(s)
Cadherins/genetics , Germ-Line Mutation , Heterozygote , Stomach Neoplasms/genetics , Antigens, CD , Breast Neoplasms/diagnosis , Breast Neoplasms/epidemiology , Breast Neoplasms/genetics , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/genetics , DNA Mutational Analysis , Early Detection of Cancer/methods , Female , Genetic Counseling , Genetic Testing/methods , Humans , Population Surveillance , Practice Guidelines as Topic , Pregnancy , Stomach Neoplasms/diagnosis , Stomach Neoplasms/epidemiology
17.
Lancet Oncol ; 16(2): e60-70, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25638682

ABSTRACT

Familial gastric cancer comprises at least three major syndromes: hereditary diffuse gastric cancer, gastric adenocarcinoma and proximal polyposis of the stomach, and familial intestinal gastric cancer. The risk of development of gastric cancer is high in families affected b-y these syndromes, but only hereditary diffuse gastric cancer is genetically explained (caused by germline alterations of CDH1, which encodes E-cadherin). Gastric cancer is also associated with a range of several cancer-associated syndromes with known genetic causes, such as Lynch, Li-Fraumeni, Peutz-Jeghers, hereditary breast-ovarian cancer syndromes, familial adenomatous polyposis, and juvenile polyposis. We present contemporary knowledge on the genetics, pathogenesis, and clinical features of familial gastric cancer, and discuss research and technological developments, which together are expected to open avenues for new genetic testing approaches and novel therapeutic strategies.


Subject(s)
Genetic Predisposition to Disease , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Humans , Stomach Neoplasms/therapy
18.
Hum Mutat ; 36(11): 1029-33, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26123647

ABSTRACT

Nonsyndromic orofacial cleft (NSOFC) is a complex disease of still unclear genetic etiology. To investigate the contribution of rare epithelial cadherin (CDH1) gene variants to NSOFC, we target sequenced 221 probands. Candidate variants were evaluated via in vitro, in silico, or segregation analyses. Three probably pathogenic variants (c.760G>A [p.Asp254Asn], c.1023T>G [p.Tyr341*], and c.2351G>A [p.Arg784His]) segregated according to autosomal dominant inheritance in four nonsyndromic cleft lip with or without cleft palate (NSCL/P) families (Lod score: 5.8 at θ = 0; 47% penetrance). A fourth possibly pathogenic variant (c.387+5G>A) was also found, but further functional analyses are needed (overall prevalence of CDH1 candidate variants: 2%; 15.4% among familial cases). CDH1 mutational burden was higher among probands from familial cases when compared to that of controls (P = 0.002). We concluded that CDH1 contributes to NSCL/P with mainly rare, moderately penetrant variants, and CDH1 haploinsufficiency is the likely etiological mechanism.


Subject(s)
Brain/abnormalities , Cadherins/genetics , Cleft Lip/genetics , Cleft Palate/genetics , Genetic Variation , Alleles , Amino Acid Substitution , Animals , Antigens, CD , Cadherins/chemistry , Cell Line , Cleft Lip/diagnosis , Cleft Palate/diagnosis , DNA Mutational Analysis , Genotype , Germ-Line Mutation , Humans , Mutation , Open Reading Frames , Penetrance
19.
Int J Cancer ; 136(4): 844-53, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-24948044

ABSTRACT

We have previously shown that gastrointestinal cancers display similar epigenetic aberrations. In a recent study, we identified frequently methylated genes for cholangiocarcinoma (CDO1, DCLK1, SFRP1 and ZSCAN18), where one of these genes, DCLK1, was also confirmed to be highly methylated in colorectal cancer. The aim of the present study was to determine whether these four genes, in addition to one gene found to be methylated in colon cancer cell lines (ZNF331), are commonly methylated across gastrointestinal malignancies, as well as explore their role as potential biomarkers. Quantitative methylation specific PCR (qMSP) of colorectal cancer (n=164) and normal colorectal mucosa (n=106) samples showed that all genes were frequently methylated in colorectal cancer (71-92%) with little or no methylation in normal mucosa (0-3%). Methylation of minimum two of these five genes identified 95% of the tumors with a specificity of 98%, and an area under the receiver operating characteristics curve (AUC) of 0.98. For gastric (n=25) and pancreatic (n=20) cancer, the same panel detected 92% and 90% of the tumors, respectively. Seventy-four cancer cell lines were further analyzed by qMSP and real time RT-PCR. In addition to the previously reported DCLK1, a high negative correlation between promoter DNA methylation and gene expression was observed for CDO1, ZNF331 and ZSCAN18. In conclusion, the high methylation frequency of these genes in colorectal- as well as in gastric-, pancreatic- and bile duct cancer confirmed an epigenetic similarity between gastrointestinal cancer types, and simultaneously demonstrated their potential as biomarkers, particularly for colorectal cancer detection.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Cysteine Dioxygenase/genetics , DNA Methylation , DNA-Binding Proteins/genetics , Neoplasm Proteins/genetics , Adult , Aged , Aged, 80 and over , Area Under Curve , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Bile Ducts, Intrahepatic/metabolism , Biomarkers, Tumor/metabolism , Case-Control Studies , Cell Line, Tumor , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Colorectal Neoplasms/metabolism , Cysteine Dioxygenase/metabolism , DNA-Binding Proteins/metabolism , Gene Expression , Humans , Middle Aged , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Promoter Regions, Genetic , ROC Curve , Sequence Analysis, DNA , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Young Adult
20.
Cancer Metastasis Rev ; 33(4): 1081-94, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25332147

ABSTRACT

Hereditary diffuse gastric cancer is an autosomic dominant syndrome associated with E-cadherin protein (CDH1) gene germline mutations. Clinical criteria for genetic screening were revised in 2010 by the International Gastric Cancer Linkage Consortium at the Cambridge meeting. About 40 % of families fulfilling clinical criteria for this inherited disease present deleterious CDH1 germline mutations. Lobular breast cancer is a neoplastic condition associated with hereditary diffuse gastric cancer syndrome. E-cadherin constitutional mutations have been described in both settings, in gastric and breast cancers. The management of CDH1 asymptomatic mutation carriers requires a multidisciplinary approach; the only life-saving procedure is the prophylactic total gastrectomy after thorough genetic counselling. Several prophylactic gastrectomies have been performed to date; conversely, no prophylactic mastectomies have been described in CDH1 mutant carriers. However, the recent discovery of novel germline alterations in pedigree clustering only for lobular breast cancer opens up a new debate in the management of these individuals. In this critical review, we describe the clinical management of CDH1 germline mutant carriers providing specific recommendations for genetic counselling, clinical criteria, surveillance and/ or prophylactic surgery.


Subject(s)
Breast Neoplasms/genetics , Cadherins/genetics , Genetic Predisposition to Disease , Stomach Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Female , Genetic Testing , Genetic Therapy , Germ-Line Mutation , Heterozygote , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery
SELECTION OF CITATIONS
SEARCH DETAIL