Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Hum Genet ; 24(10): 1430-5, 2016 10.
Article in English | MEDLINE | ID: mdl-27049303

ABSTRACT

Tooth development is controlled by the same processes that regulate formation of other ectodermal structures. Mutations in the genes underlying these processes may cause ectodermal dysplasia, including severe absence of primary or permanent teeth. Four consanguineous Palestinian families presented with oligodontia and hair and skin features of ectodermal dysplasia. Appearance of ectodermal dysplasia was consistent with autosomal recessive inheritance. Exome sequencing followed by genotyping of 56 informative relatives in the 4 families suggests that the phenotype is due to homozygosity for KREMEN1 p.F209S (c.626 T>C) on chromosome 22 at g.29,521,399 (hg19). The variant occurs in the highly conserved extracellular WSC domain of KREMEN1, which is known to be a high affinity receptor of Dickkopf-1, a component of the Dickkopf-Kremen-LRP6 complex, and a potent regulator of Wnt signaling. The Wnt signaling pathway is critical to development of ectodermal structures. Mutations in WNT10A, LRP6, EDA, and other genes in this pathway lead to tooth agenesis with or without other ectodermal anomalies. Our results implicate KREMEN1 for the first time in a human disorder and provide additional details on the role of the Wnt signaling in ectodermal and dental development.


Subject(s)
Anodontia/genetics , Ectodermal Dysplasia/genetics , Membrane Proteins/genetics , Mutation , Wnt Signaling Pathway , Adolescent , Anodontia/diagnosis , Child , Chromosomes, Human, Pair 22/genetics , Ectodermal Dysplasia/diagnosis , Exome , Female , Genes, Recessive , Humans , Male , Pedigree , Syndrome
2.
Breast Care (Basel) ; 6(4): 265-272, 2011.
Article in English | MEDLINE | ID: mdl-22135624

ABSTRACT

SUMMARY: About 10-25% of breast cancer patients achieve a pathologically confirmed complete response after neoadjuvant chemotherapy. Tissue samples of pretreatment core biopsies are a valuable resource for translational research aiming towards predictive biomarkers for selecting patients who are likely to benefit from neoadjuvant therapy. The German Breast Group (GBG) and the AGO-B Group (AGO = Working Group Gynecological Oncology) have extensive experience in conducting neoadjuvant clinical trials. Technologies as immunohistochemistry on tissue microarrays and standardized reverse transcription-polymerase chain reaction (RT-PCR) approaches on formalin-fixed paraffin-embedded samples allow high-throughput investigation of protein and mRNA biomarkers. With these approaches, we could demonstrate that molecular tumor subtypes and immunological infiltrates are valuable and independent predictors of therapy response. New biomarkers such as poly(ADPribose) polymerase (PARP) might be useful for the prediction of response to conventional and new targeted therapies. This review summarizes current research projects focusing on biomarker discovery in the neoadjuvant setting.

3.
J Mol Med (Berl) ; 87(1): 99-112, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18931829

ABSTRACT

L1 cell adhesion molecule (L1CAM) is a transmembrane cell adhesion molecule initially defined as a promigratory molecule in the developing nervous system that appears to be also expressed in some endothelial cells. However, little is known about the functional role of L1CAM on endothelial cells. We observed that L1CAM expression was selectively enhanced on endothelium associated with pancreatic adenocarcinoma in situ and on cultured pancreatic tumor-derived endothelial cells in vitro. L1CAM expression of endothelial cells could be augmented by incubation with immunomodulatory cytokines such as tumor necrosis factor alpha, interferon gamma, or transforming growth factor beta 1. Antibodies to L1CAM and the respective ligand neuropilin-1 blocked tube formation and stromal cell-derived factor 1beta induced transmigration of tumor endothelial cells in vitro. L1CAM expression on tumor-derived-endothelial cells enhanced Panc1 carcinoma cell adhesion to endothelial cell monolayers and transendothelial migration. Our data demonstrate a functional role of L1CAM expression on tumor endothelium that could favor metastasis and angiogenesis during tumor progression.


Subject(s)
Adenocarcinoma/metabolism , Cell Movement , Endothelium/metabolism , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Neural Cell Adhesion Molecule L1/biosynthesis , Pancreatic Neoplasms/metabolism , Adenocarcinoma/pathology , Antibodies, Neoplasm/pharmacology , Cytokines/pharmacology , Endothelium/pathology , Humans , Neoplasm Metastasis , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neuropilin-1/antagonists & inhibitors , Neuropilin-1/metabolism , Pancreatic Neoplasms/pathology , Tumor Cells, Cultured
4.
Gynecol Oncol ; 107(3): 563-71, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17900673

ABSTRACT

OBJECTIVE: CD24 is an established marker for poor prognosis in ovarian and other carcinomas. Acquisition of cytoplasmic CD24, as opposed to membranous expression, has been correlated with a higher invasiveness of tumor cells. Exosomes are small vesicles of endosomal origin that are often secreted by tumor cells. Given the emerging role of exosomes in tumor progression, we investigated whether cytoplasmic CD24 expression is correlated with the secretion of CD24 in exosomes. METHODS: We used CD24 transfected carcinoma cell lines, ovarian carcinoma cell lines and malignant ascites fluid of ovarian carcinoma patients. Exosomes were isolated via ultracentrifugation and sucrose density fractionation and subsequently examined by Western blot analysis and gelatine zymography. In tissue sections CD24 was detected by immunohistochemical staining. RESULTS: We show that CD24 is released by transfected as well as endogenously expressing cells in vesicles that represent exosomes. CD24 was also identified in exosomes isolated from ascites fluid of ovarian carcinoma patients. In 16 ovarian carcinomas analyzed no correlation between CD24 in tumor tissue sections and the appearance of CD24 in exosomes was detected. Furthermore, we provide evidence that the epithelial cell adhesion molecule (EpCAM), known to be overexpressed in ovarian carcinomas, is secreted in exosomes. The ascites exosomes contain gelatinolytic enzymes. CONCLUSIONS: Our study identifies CD24 and EpCAM as cargo proteins of exosomes of cultured cell lines and malignant ascites. The exosome-associated proteolytic activity in the tumor vicinity might augment tumor invasion into the stroma.


Subject(s)
Antigens, Neoplasm/metabolism , CD24 Antigen/metabolism , Cell Adhesion Molecules/metabolism , Ovarian Neoplasms/metabolism , Secretory Vesicles/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Ascites/metabolism , CD24 Antigen/biosynthesis , CD24 Antigen/genetics , Cell Line, Tumor , Epithelial Cell Adhesion Molecule , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Transfection
5.
Int J Cancer ; 120(12): 2625-34, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17294447

ABSTRACT

Expression of the neural cell adhesion molecule (NCAM) on malignant cells of neuroendocrine, epithelial and hematopoeitic origin has been reported, but its role for tumor cell recognition by the immune system remained uncertain so far. We have studied the cytotoxicity of the natural killer (NK) cell line NK92 and polyclonal NK cells from different donors, against NCAM-deficient and NCAM-transfected tumors. While the pancreatic carcinoma PANC-1 and the glioblastoma T98G showed no enhanced susceptibility to NK lysis after NCAM transfection, de novo NCAM expression in HeLa cervical carcinoma, SHEP neuroblastoma and the multiple myeloma lines RPMI-8226 and LP-1 was associated with significantly decreased lysis by NK cells. Binding of an NCAM-specific monoclonal antibody to NCAM-positive target cells was able to reverse the reduced lysis susceptibility. Conjugate formation of NCAM-expressing tumor cells with NK cells was blocked and could be restored by anti-NCAM. NK cell-expressed NCAM molecules which might engage in homotypic cis- or trans-interactions had no apparent inhibitory function. The known cis-ligands of NCAM, heparan sulfate proteoglycan and L1-CAM, were also not directly involved in NK inhibition. ICAM-1 mRNA and cell surface expression was downmodulated in NCAM-transfected HeLa cells. ICAM-1 is involved in killer cell immune synapse formation. Its downmodulation may therefore contribute to the reduced lysis of NCAM-expressing target cells. We conclude that aberrant expression of NCAM on tumor cells of different histogenetic origin can lead to inhibition of target cell recognition and lysis by NK cells.


Subject(s)
Cell Adhesion Molecules, Neuronal/immunology , Cytotoxicity, Immunologic/immunology , Killer Cells, Natural/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , CHO Cells , Cell Adhesion Molecules, Neuronal/genetics , Cell Line , Cell Line, Tumor , Clone Cells , Cricetinae , Cricetulus , Cytotoxicity Tests, Immunologic , Cytotoxicity, Immunologic/drug effects , Dose-Response Relationship, Drug , Gene Expression , HeLa Cells , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Killer Cells, Natural/metabolism , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection
6.
Gynecol Oncol ; 104(2): 461-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17030349

ABSTRACT

OBJECTIVE: Apoptosis resistance is a hallmark of cancer progression, a phenomenon frequently observed in ovarian carcinoma. We reported previously, that L1 adhesion molecule (CD171) is overexpressed in ovarian and endometrial carcinomas and that L1 expression is a predictor of poor outcome. We investigated a possible role of L1 in apoptosis resistance. METHODS: We used L1 transfectants and ovarian carcinoma cell lines and induced apoptosis by different stimuli such as C2-ceramide, staurosporine, cisplatin or hypoxia. RESULTS: We found that cells expressing L1 are more resistant against apoptosis. In HEK293 cells, L1-expresssion leads to a sustained ERK, FAK and PAK phosphorylation. Soluble L1 only partially rescued HEK293 cells from apoptosis. Treatment with apoptotic stimuli upregulated the anti-apoptotic molecule Bcl-2 to a greater extend in HEK293 cells expressing L1. In the ovarian carcinoma cell line OVMz, the depletion of L1 by RNA interference sensitized cells for apoptosis induction. No changes in activation of ERK or FAK were observed after L1 knockdown. The selection of m130 ovarian carcinoma or SW707 colon carcinoma cells with cisplatin leads to upregulated expression of L1. CONCLUSIONS: Our results suggest a link between L1 expression and chemoresistance of ovarian carcinomas. Upregulation of L1 after cisplatin treatment might indicate a more malignant tumor phenotype given the established role of L1 in cell motility and invasion.


Subject(s)
Apoptosis/physiology , Neural Cell Adhesion Molecule L1/physiology , Ovarian Neoplasms/pathology , Animals , Apoptosis/drug effects , Apoptosis/genetics , CHO Cells , Cell Line , Cell Line, Tumor , Cisplatin/pharmacology , Cricetinae , Cricetulus , Female , Humans , Neural Cell Adhesion Molecule L1/genetics , Ovarian Neoplasms/genetics , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Staurosporine/pharmacology , Transfection
7.
Cancer Lett ; 239(2): 212-26, 2006 Aug 08.
Article in English | MEDLINE | ID: mdl-16377081

ABSTRACT

The progression of ovarian cancer is driven by a variety of cellular factors that are incompletely understood. Binding of tumor cells to normal cells and to soluble factors influence tumor growth, angiogenesis and the stimulation of vascular permeability leading to ascites production. L1 adhesion molecule is overexpressed in ovarian carcinoma and is associated with bad prognosis. One receptor for L1 is Neuropilin-1 (NRP-1) that is also known as a receptor for VEGF(165). In the nervous system a complex of NRP-1 and L1 transmits signals by the neurorepellant Sem3A that is critical for the control of neurite outgrowth. NRP-1 has also been detected in human carcinomas but its function remains unknown. Here, we have examined NRP-1 expression in ovarian carcinoma cell lines and tissue. We report that little NRP-1 protein was detected in primary ovarian carcinoma tissues or established cell lines although mRNA for soluble and transmembrane NRP-1 were detected by RT-PCR. Instead, we observed strong expression of NRP-1 in mesothelial cells, which form the lining of the peritoneum. NRP-1 could serve as an isolation marker for primary mesothelial cells present in ascites fluid. We demonstrate that ovarian cancer cells expressing L1 can bind to NRP-1 overexpressing cells and mesothelial cells. Likewise, soluble L1 isolated from ascites of patients or produced as a fusion protein could bind to NRP-1 overexpressing cells and a direct interaction was demonstrated at the protein level. These findings suggest that L1 can support the binding of ovarian carcinoma cells to mesothelial cells via NRP-1. The L1-NRP-1 binding pathway could contribute to the growth of ovarian carcinomas and to reciprocal signalling between mesothelial cells and tumors.


Subject(s)
Neural Cell Adhesion Molecule L1/metabolism , Neuropilin-1/metabolism , Ovarian Neoplasms/metabolism , Base Sequence , Blotting, Western , Cell Line, Tumor , DNA Primers , Female , Humans , Microscopy, Fluorescence , Ovarian Neoplasms/pathology , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction
8.
Int J Cancer ; 115(4): 658-65, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15704102

ABSTRACT

L1 is a neural cell adhesion molecule involved in cell migration, axon growth and guidance. Recent data have shown that L1 is overexpressed in ovarian and endometrial tumors and is associated with bad prognosis. How L1 promotes tumor progression is presently unknown. Here we show that L1 expression is predominantly confined to the invasive front of ovarian carcinomas. Overexpression of L1 in carcinoma cell lines by adenovirus-mediated gene transfer enhanced the haptotactic cell migration on extracellular matrix proteins. Expression of L1 augmented tumor growth of carcinomas xenografted in nonobese diabetic/severe combined immunodeficient mice (NOD/SCID). A recent report has demonstrated L1-dependent upregulation of beta3 integrin involving activation of the extracellular signal-regulated kinase (erk) pathway. We find that L1 and beta3 integrin are not coexpressed in ovarian carcinoma tissues. Overexpression of L1 did not upregulate beta3 integrin in ovarian carcinoma cell lines but could do so in HEK293 cells. Our results suggest that L1 could drive progression by enhancing cell migration and tumor growth but that L1 dependent and erk-regulated gene expression requires cell-type specific elements.


Subject(s)
Integrin beta3/genetics , Neural Cell Adhesion Molecule L1/genetics , Ovarian Neoplasms/genetics , Animals , Cell Division , Cell Line , Cell Line, Tumor , Cell Movement , Female , Humans , Kidney , Mice , Mice, Inbred NOD , Mice, SCID , Neural Cell Adhesion Molecule L1/metabolism , Ovarian Neoplasms/pathology , RNA, Small Interfering/genetics , Transfection , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...