Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Exp Clin Cancer Res ; 41(1): 285, 2022 Sep 26.
Article in English | MEDLINE | ID: mdl-36163066

ABSTRACT

BACKGROUND: Gasdermin B (GSDMB) over-expression promotes poor prognosis and aggressive behavior in HER2 breast cancer by increasing resistance to therapy. Decoding the molecular mechanism of GSDMB-mediated drug resistance is crucial to identify novel effective targeted treatments for HER2/GSDMB aggressive tumors. METHODS: Different in vitro approaches (immunoblot, qRT-PCR, flow cytometry, proteomic analysis, immunoprecipitation, and confocal/electron microscopy) were performed in HER2 breast and gastroesophageal carcinoma cell models. Results were then validated using in vivo preclinical animal models and analyzing human breast and gastric cancer samples. RESULTS: GSDMB up-regulation renders HER2 cancer cells more resistant to anti-HER2 agents by promoting protective autophagy. Accordingly, the combination of lapatinib with the autophagy inhibitor chloroquine increases the therapeutic response of GSDMB-positive cancers in vitro and in zebrafish and mice tumor xenograft in vivo models. Mechanistically, GSDMB N-terminal domain interacts with the key components of the autophagy machinery LC3B and Rab7, facilitating the Rab7 activation during pro-survival autophagy in response to anti-HER2 therapies. Finally, we validated these results in clinical samples where GSDMB/Rab7/LC3B co-expression associates significantly with relapse in HER2 breast and gastric cancers. CONCLUSION: Our findings uncover for the first time a functional link between GSDMB over-expression and protective autophagy in response to HER2-targeted therapies. GSDMB behaves like an autophagy adaptor and plays a pivotal role in modulating autophagosome maturation through Rab7 activation. Finally, our results provide a new and accessible therapeutic approach for HER2/GSDMB + cancers with adverse clinical outcome.


Subject(s)
Breast Neoplasms , Receptor, ErbB-2 , Animals , Autophagy , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Chloroquine/pharmacology , Drug Resistance, Neoplasm , Female , Humans , Lapatinib/pharmacology , Mice , Neoplasm Recurrence, Local , Proteomics , Receptor, ErbB-2/genetics , Zebrafish
2.
Front Cell Dev Biol ; 10: 813929, 2022.
Article in English | MEDLINE | ID: mdl-35281099

ABSTRACT

Gasdermins (GSDM) genes play complex roles in inflammatory diseases and cancer. Gasdermin-B (GSDMB) is frequently upregulated in human cancers, especially in HER2-amplified breast carcinomas, and can promote diverse pro-tumor functions (invasion, metastasis, therapy-resistance). In particular, the GSDMB shortest translated variant (isoform 2; GSDMB2) increases aggressive behavior in breast cancer cells. Paradoxically, GSDMB can also have tumor suppressor (cell death induction) effects in specific biological contexts. However, whether GSDMB has inherent oncogenic, or tumor suppressor function in vivo has not been demonstrated yet in preclinical mouse models, since mice lack GSDMB orthologue. Therefore, to decipher GSDMB cancer functions in vivo we first generated a novel knock-in mouse model (R26-GB2) ubiquitously expressing human GSDMB2. The comprehensive histopathological analysis of multiple tissues from 75 animals showed that nucleus-cytoplasmic GSDMB2 expression did not clearly affect the overall frequency nor the histology of spontaneous neoplasias (mostly lung carcinomas), but associated with reduced incidence of gastric tumors, compared to wildtype animals. Next, to assess specifically the GSDMB2 roles in breast cancer, we generated two additional double transgenic mouse models, that co-express GSDMB2 with either the HER2/NEU oncogene (R26-GB2/MMTV-NEU mice) or the Polyoma middle-T antigen (R26-GB2/MMTV-PyMT) in breast tumors. Consistent with the pro-tumor effect of GSDMB in HER2+ human breast carcinomas, R26-GB2/MMTV-NEU GSDMB2-positive mice have double breast cancer incidence than wildtype animals. By contrast, in the R26-GB2/MMTV-PyMT model of fast growing and highly metastatic mammary tumors, GSDMB2 expression did not significantly influence cancer development nor metastatic potential. In conclusion, our data prove that GSDMB2 in vivo pro-tumor effect is evidenced only in specific biological contexts (in concert with the HER2 oncogene), while GSDMB2 alone does not have overall intrinsic oncogenic potential in genetically modified mice. Our novel models are useful to identify the precise stimuli and molecular mechanisms governing GSDMB functions in neoplasias and can be the basis for the future development of additional tissue-specific and context-dependent cancer models.

3.
Oncogene ; 41(13): 1835-1850, 2022 03.
Article in English | MEDLINE | ID: mdl-35145232

ABSTRACT

Analyzing different tumor regions by next generation sequencing allows the assessment of intratumor genetic heterogeneity (ITGH), a phenomenon that has been studied widely in some tumor types but has been less well explored in endometrial carcinoma (EC). In this study, we sought to characterize the spatial and temporal heterogeneity of 9 different ECs using whole-exome sequencing, and by performing targeted sequencing validation of the 42 primary tumor regions and 30 metastatic samples analyzed. In addition, copy number alterations of serous carcinomas were assessed by comparative genomic hybridization arrays. From the somatic mutations, identified by whole-exome sequencing, 532 were validated by targeted sequencing. Based on these data, the phylogenetic tree reconstructed for each case allowed us to establish the tumors' evolution and correlate this to tumor progression, prognosis, and the presence of recurrent disease. Moreover, we studied the genetic landscape of an ambiguous EC and the molecular profile obtained was used to guide the selection of a potential personalized therapy for this patient, which was subsequently validated by preclinical testing in patient-derived xenograft models. Overall, our study reveals the impact of analyzing different tumor regions to decipher the ITGH in ECs, which could help make the best treatment decision.


Subject(s)
Endometrial Neoplasms , Genetic Heterogeneity , Clonal Evolution/genetics , Comparative Genomic Hybridization , DNA Copy Number Variations/genetics , Endometrial Neoplasms/genetics , Female , Humans , Mutation , Phylogeny
4.
Cells ; 9(5)2020 05 14.
Article in English | MEDLINE | ID: mdl-32423054

ABSTRACT

BACKGROUND: Recent studies showed a relevant role of hematogenous spread in ovarian cancer and the interest of circulating tumor cells (CTCs) monitoring as a prognosis marker. The aim of the present study was the characterization of CTCs from ovarian cancer patients, paying special attention to cell plasticity characteristics to better understand the biology of these cells. METHODS: CTCs isolation was carried out in 38 patients with advanced high-grade serous ovarian cancer using in parallel CellSearch and an alternative EpCAM-based immunoisolation followed by RT-qPCR analysis to characterize these cells. RESULTS: Epithelial CTCs were found in 21% of patients, being their presence higher in patients with extraperitoneal metastasis. Importantly, this population was characterized by the expression of epithelial markers as MUC1 and CK19, but also by genes associated with mesenchymal and more malignant features as TIMP1, CXCR4 and the stem markers CD24 and CD44. In addition, we evidenced the relevance of TIMP1 expression to promote tumor proliferation, suggesting its interest as a therapeutic target. CONCLUSIONS: Overall, we evidenced the utility of the molecular characterization of EpCAM+ CTCs from advanced ovarian cancer patients to identify biomarkers with potential applicability for disseminated disease detection and as therapeutic targets such as TIMP1.


Subject(s)
Molecular Targeted Therapy , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/enzymology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Cell Plasticity , Cell Proliferation , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Middle Aged , Neoplasm Invasiveness , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Xenograft Model Antitumor Assays , Young Adult , Zebrafish
6.
Neoplasia ; 21(11): 1073-1084, 2019 11.
Article in English | MEDLINE | ID: mdl-31734628

ABSTRACT

The role of prostaglandin (PG) F2α has been scarcely studied in cancer. We have identified a new function for PGF2α in ovarian cancer, stimulating the production of Prostate Transmembrane Protein, Androgen Induced 1 (PMEPA1). We show that this induction increases cell plasticity and proliferation, enhancing tumor growth through PMEPA1. Thus, PMEPA1 overexpression in ovarian carcinoma cells, significantly increased cell proliferation rates, whereas PMEPA1 silencing decreased proliferation. In addition, PMEPA1 overexpression buffered TGFß signaling, via reduction of SMAD-dependent signaling. PMEPA1 overexpressing cells acquired an epithelial morphology, associated with higher E-cadherin expression levels while ß-catenin nuclear translocation was inhibited. Notwithstanding, high PMEPA1 levels also correlated with epithelial to mesenchymal transition markers, such as vimentin and ZEB1, allowing the cells to take advantage of both epithelial and mesenchymal characteristics, gaining in cell plasticity and adaptability. Interestingly, in mouse xenografts, PMEPA1 overexpressing ovarian cells had a clear survival and proliferative advantage, resulting in higher metastatic capacity, while PMEPA1 silencing had the opposite effect. Furthermore, high PMEPA1 expression in a cohort of advanced ovarian cancer patients was observed, correlating with E-cadherin expression. Most importantly, high PMEPA1 mRNA levels were associated with lower patient survival.

7.
Sci Rep ; 8(1): 13038, 2018 08 29.
Article in English | MEDLINE | ID: mdl-30158566

ABSTRACT

Although Ras genes are frequently mutated in human tumors, these mutations are uncommon in breast cancer. However, many breast tumors show evidences of Ras pathway activation. In this manuscript, we have analyzed and characterized mouse mammary tumors generated by random Sleeping Beauty transposon mutagenesis and identify ERAS -a member of the RAS family silenced in adult tissues- as a new gene involved in progression and malignancy of breast cancer. Forced expression of ERAS in human non-transformed mammary gland cells induces a process of epithelial-to-mesenchymal transition and an increase in stem cells markers; these changes are mediated by miR-200c downregulation. ERAS expression in human tumorigenic mammary cells leads to the generation of larger and less differentiated tumors in xenotransplant experiments. Immunohistochemical, RT-qPCR and bioinformatics analysis of human samples show that ERAS is aberrantly expressed in 8-10% of breast tumors and this expression is associated with distant metastasis and reduced metastasis-free survival. In summary, our results reveal that inappropriate activation of ERAS may be important in the development of a subset of breast tumors. These findings open the possibility of new specific treatments for this subset of ERAS-expressing tumors.


Subject(s)
Breast Neoplasms/physiopathology , Oncogene Protein p21(ras)/metabolism , Animals , Breast Neoplasms/pathology , Carcinogenesis , Cell Differentiation , Cells, Cultured , Epithelial Cells/physiology , Epithelial-Mesenchymal Transition , Humans , Mice , Neoplasm Transplantation , Neoplasms, Experimental/pathology , Neoplasms, Experimental/physiopathology , Oncogene Protein p21(ras)/genetics , Transplantation, Heterologous
8.
Mod Pathol ; 30(1): 134-145, 2017 01.
Article in English | MEDLINE | ID: mdl-27586201

ABSTRACT

Endometrial cancer is the most common cancer of the female genital tract in developed countries. Although the majority of endometrial cancers are diagnosed at early stages and the 5-year overall survival is around 80%, early detection of these tumors is crucial to improve the survival of patients given that the advanced tumors are associated with a poor outcome. Furthermore, correct assessment of the pre-clinical diagnosis is decisive to guide the surgical treatment and management of the patient. In this sense, the potential of targeted genetic sequencing of uterine aspirates has been assessed as a pre-operative tool to obtain reliable information regarding the mutational profile of a given tumor, even in samples that are not histologically classifiable. A total of 83 paired samples were sequenced (uterine aspirates and hysterectomy specimens), including 62 endometrioid and non-endometrioid tumors, 10 cases of atypical hyperplasia and 11 non-cancerous endometrial disorders. Even though diagnosing endometrial cancer based exclusively on genetic alterations is currently unfeasible, mutations were mainly found in uterine aspirates from malignant disorders, suggesting its potential in the near future for supporting the standard histologic diagnosis. Moreover, this approach provides the first evidence of the high intra-tumor genetic heterogeneity associated with endometrial cancer, evident when multiple regions of tumors are analyzed from an individual hysterectomy. Notably, the genetic analysis of uterine aspirates captures this heterogeneity, solving the potential problem of incomplete genetic characterization when a single tumor biopsy is analyzed.


Subject(s)
Carcinoma, Endometrioid/diagnosis , Carcinosarcoma/diagnosis , Endometrial Neoplasms/diagnosis , Uterus/pathology , Aged , Aged, 80 and over , Carcinoma, Endometrioid/genetics , Carcinoma, Endometrioid/pathology , Carcinosarcoma/genetics , Carcinosarcoma/pathology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Humans , Middle Aged , Mutation
9.
Int J Cancer ; 140(7): 1551-1563, 2017 04 01.
Article in English | MEDLINE | ID: mdl-27997699

ABSTRACT

In developed countries, endometrial carcinoma is the most common cancer that affects the female genital tract. Endometrial carcinoma is divided into two main histological types, type I or endometrioid and type II or non-endometrioid, each of which have characteristic, although not exclusive, molecular alterations and mutational profiles. Nevertheless, information about the implication and relevance of some of these genes in this disease is lacking. We sought here to identify new recurrently mutated genes in endometrioid cancers that play a role in tumourigenesis and that influence the clinical outcome. We focused on low-grade, non-ultramutated tumours as these tumours have a worse prognosis than the ultramutated POLE-positive endometrioid endometrial carcinomas (EECs). We performed exome-sequencing of 11 EECs with matched normal tissue and subsequently validated 15 candidate genes in 76 samples. For the first time, we show that mutations in chromatin remodelling-related genes (KMT2D, KMT2C, SETD1B and BCOR) and in DNA-repair-related genes (BRCA1, BRCA2, RAD50 and CHD4) are frequent in this subtype of endometrial cancer. The alterations to these genes occurred with frequencies ranging from 35.5% for KMT2D to 10.5% for BRCA1 and BCOR, with some showing a tendency toward co-occurrence (RAD50-KMT2D and RAD50-SETD1B). All these genes harboured specific mutational hotspots. In addition, the mutational status of KMT2C, KMT2D and SETD1B helps to predict the degree of myometrial invasion, a critical prognostic feature. These results highlight the possible implication of these genes in this disease, creating opportunities for new therapeutic approaches.


Subject(s)
Carcinoma, Endometrioid/pathology , Chromatin Assembly and Disassembly , Chromatin/chemistry , DNA Repair , Endometrial Neoplasms/pathology , Mutation , Carcinoma, Endometrioid/genetics , Cell Line, Tumor , Computational Biology , DNA Mutational Analysis , Endometrial Neoplasms/genetics , Exome , Female , Genetic Predisposition to Disease , Humans , Immunohistochemistry , Microsatellite Instability , Mutation, Missense , Myometrium/metabolism , Myometrium/pathology , Prognosis
10.
Oncotarget ; 7(37): 60110-60122, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27507050

ABSTRACT

The truncated somatostatin receptor sst5TMD4 is associated with poor prognosis in breast cancer and increases breast cancer cell malignancy. Here, we examined the cellular/molecular mechanisms underlying this association, aiming to identify new molecular tools to improve diagnosis, prognosis or therapy. A gene expression array comparing sst5TMD4 stably-transfected MCF-7 cells and their controls (empty-plasmid) revealed the existence of profound alterations in the expression of genes involved in key tumoral processes, such as cell survival or angiogenesis. Moreover, sst5TMD4-overexpressing MCF-7 and MDA-MB-231 cells demonstrated increased expression/production of pro-angiogenic factors and enhanced capacity to form mammospheres. Consistently, sst5TMD4-expressing MCF-7 cells induced xenografted tumors with higher VEGF levels and elevated number of blood vessels. Importantly, sst5TMD4 was expressed in a subset of breast cancers, where it correlated with angiogenic markers, lymphatic metastasis, and reduced disease-free survival. These results, coupled to our previous data, support a relevant role of sst5TMD4 in the angiogenic process and reinforce the role of sst5TMD4 in breast cancer malignancy and metastatic potential, supporting its possible utility to develop new molecular biomarkers and drug therapies for these tumors.


Subject(s)
Breast Neoplasms/genetics , Mutation , Neovascularization, Pathologic/genetics , Receptors, Somatostatin/genetics , Adult , Aged , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Lymphatic Metastasis , MCF-7 Cells , Mice, Nude , Middle Aged , Neovascularization, Pathologic/metabolism , Receptors, Somatostatin/metabolism , Transplantation, Heterologous
11.
Oncotarget ; 7(35): 56295-56308, 2016 Aug 30.
Article in English | MEDLINE | ID: mdl-27462779

ABSTRACT

Around, 30-40% of HER2-positive breast cancers do not show substantial clinical benefit from the targeted therapy and, thus, the mechanisms underlying resistance remain partially unknown. Interestingly, ERBB2 is frequently co-amplified and co-expressed with neighbour genes that may play a relevant role in this cancer subtype. Here, using an in silico analysis of data from 2,096 breast tumours, we reveal a significant correlation between Gasdermin B (GSDMB) gene (located 175 kilo bases distal from ERBB2) expression and the pathological and clinical parameters of poor prognosis in HER2-positive breast cancer. Next, the analysis of three independent cohorts (totalizing 286 tumours) showed that approximately 65% of the HER2-positive cases have GSDMB gene amplification and protein over-expression. Moreover, GSDMB expression was also linked to poor therapeutic responses in terms of lower relapse free survival and pathologic complete response as well as positive lymph node status and the development of distant metastasis under neoadjuvant and adjuvant treatment settings, respectively. Importantly, GSDMB expression promotes survival to trastuzumab in different HER2-positive breast carcinoma cells, and is associated with trastuzumab resistance phenotype in vivo in Patient Derived Xenografts. In summary, our data identifies the ERBB2 co-amplified and co-expressed gene GSDMB as a critical determinant of poor prognosis and therapeutic response in HER2-positive breast cancer.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Neoplasm Recurrence, Local/genetics , Receptor, ErbB-2/antagonists & inhibitors , Adult , Aged , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Carcinoma, Ductal, Breast/mortality , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/therapy , Chemotherapy, Adjuvant , Disease-Free Survival , Drug Resistance, Neoplasm , Female , Follow-Up Studies , Gene Amplification , Humans , Lymphatic Metastasis , Mice , Mice, Nude , Middle Aged , Neoadjuvant Therapy , Neoplasm Proteins/metabolism , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/therapy , Prognosis , Receptor, ErbB-2/metabolism , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Xenograft Model Antitumor Assays
12.
BMC Cancer ; 15: 940, 2015 Nov 30.
Article in English | MEDLINE | ID: mdl-26620706

ABSTRACT

BACKGROUND: High grade serous ovarian cancer is characterised by high initial response to chemotherapy but poor outcome in the long term due to acquired resistance. One of the main genetic features of this disease is TP53 mutation. The majority of TP53 mutated tumors harbor missense mutations in this gene, correlated with p53 accumulation. TP53 null tumors constitute a specific subgroup characterised by nonsense, frameshift or splice-site mutations associated to complete absence of p53 expression. Different studies show that this kind of tumors may have a worse prognosis than other TP53 mutated HGSC. METHODS: In this study, we sought to characterise the intra-tumor heterogeneity of a TP53 null HGSC consisting of six primary tumor samples, two intra-pelvic and four extra-pelvic recurrences using exome sequencing and comparative genome hybridisation. RESULTS: Significant heterogeneity was found among the different tumor samples, both at the mutational and copy number levels. Exome sequencing identified 102 variants, of which only 42 were common to all three samples; whereas 7 of the 18 copy number changes found by CGH analysis were presented in all samples. Sanger validation of 20 variants found by exome sequencing in additional regions of the primary tumor and the recurrence allowed us to establish a sequence of the tumor clonal evolution, identifying those populations that most likely gave rise to recurrences and genes potentially involved in this process, like GPNMB and TFDP1. Using functional annotation and network analysis, we identified those biological functions most significantly altered in this tumor. Remarkably, unexpected functions such as microtubule-based movement and lipid metabolism emerged as important for tumor development and progression, suggesting its potential interest as therapeutic targets. CONCLUSIONS: Altogether, our results shed light on the clonal evolution of the distinct tumor regions identifying the most aggressive subpopulations and at least some of the genes that may be implicated in its progression and recurrence, and highlights the importance of considering intra-tumor heterogeneity when carrying out genetic and genomic studies, especially when these are aimed to diagnostic procedures or to uncover possible therapeutic strategies.


Subject(s)
Cystadenocarcinoma, Serous/pathology , Genetic Heterogeneity , Ovarian Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Clonal Evolution , Comparative Genomic Hybridization , Cystadenocarcinoma, Serous/genetics , Female , Genetic Variation , Humans , Membrane Glycoproteins/genetics , Middle Aged , Mutation , Neoplasm Grading , Ovarian Neoplasms/genetics , Prognosis , Sequence Analysis, DNA , Transcription Factor DP1/genetics
13.
PLoS One ; 9(3): e90099, 2014.
Article in English | MEDLINE | ID: mdl-24675552

ABSTRACT

Gasdermin B (GSDMB) belongs to the Gasdermin protein family that comprises four members (GSDMA-D). Gasdermin B expression has been detected in some tumor types such as hepatocarcinomas, gastric and cervix cancers; and its over-expression has been related to tumor progression. At least four splicing isoforms of GSDMB have been identified, which may play differential roles in cancer. However, the implication of GSDMB in carcinogenesis and tumor progression is not well understood. Here, we uncover for the first time the functional implication of GSDMB in breast cancer. Our data shows that high levels of GSDMB expression is correlated with reduced survival and increased metastasis in breast cancer patients included in an expression dataset (>1,000 cases). We demonstrate that GSDMB is upregulated in breast carcinomas compared to normal breast tissue, being the isoform 2 (GSDMB-2) the most differentially expressed. In order to evaluate the functional role of GSDMB in breast cancer two GSDMB isoforms were studied (GSDMB-1 and GSDMB-2). The overexpression of both isoforms in the MCF7 breast carcinoma cell line promotes cell motility and invasion, while its silencing in HCC1954 breast carcinoma cells decreases the migratory and invasive phenotype. Importantly, we demonstrate that both isoforms have a differential role on the activation of Rac-1 and Cdc-42 Rho-GTPases. Moreover, our data support that GSMDB-2 induces a pro-tumorigenic and pro-metastatic behavior in mouse xenograft models as compared to GSDMB-1. Finally, we observed that although both GSDMB isoforms interact in vitro with the chaperone Hsp90, only the GSDMB-2 isoform relies on this chaperone for its stability. Taken together, our results provide for the first time evidences that GSDMB-2 induces invasion, tumor progression and metastasis in MCF7 cells and that GSDMB can be considered as a new potential prognostic marker in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Neoplasm Proteins/genetics , Aged , Aged, 80 and over , Animals , Breast Neoplasms/mortality , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/mortality , Carcinoma, Ductal, Breast/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Datasets as Topic , Disease Models, Animal , Female , Gelatin/metabolism , Gene Expression , Genes, Reporter , HSP90 Heat-Shock Proteins/metabolism , Heterografts , Humans , Mice , Middle Aged , Molecular Imaging , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/metabolism , Phenotype , Prognosis , Protein Binding , Proteolysis , rho GTP-Binding Proteins/metabolism
16.
EMBO Mol Med ; 3(9): 528-44, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21732535

ABSTRACT

Basal-like breast carcinoma is characterized by the expression of basal/myoepithelial markers, undifferentiated phenotype, highly aggressive behaviour and frequent triple negative status (ESR-, PR-, Her2neu-). We have previously shown that epithelial-mesenchymal transition (EMT) occurs in basal-like breast tumours and identified Lysyl-oxidase-like 2 (LOXL2) as an EMT player and poor prognosis marker in squamous cell carcinomas. We now show that LOXL2 mRNA is overexpressed in basal-like human breast carcinomas. Breast carcinoma cell lines with basal-like phenotype show a specific cytoplasmic/perinuclear LOXL2 expression, and this subcellular distribution is significantly associated with distant metastatic incidence in basal-like breast carcinomas. LOXL2 silencing in basal-like carcinoma cells induces a mesenchymal-epithelial transition (MET) associated with a decrease of tumourigenicity and suppression of metastatic potential. Mechanistic studies indicate that LOXL2 maintains the mesenchymal phenotype of basal-like carcinoma cells by a novel mechanism involving transcriptional downregulation of Lgl2 and claudin1 and disorganization of cell polarity and tight junction complexes. Therefore, intracellular LOXL2 is a new candidate marker of basal-like carcinomas and a target to block metastatic dissemination of this aggressive breast tumour subtype.


Subject(s)
Adenocarcinoma/pathology , Adenocarcinoma/secondary , Amino Acid Oxidoreductases/metabolism , Breast Neoplasms/parasitology , Breast Neoplasms/secondary , Cell Polarity , Neoplasm Metastasis/pathology , Biomarkers , Female , Humans
17.
Adv Urol ; : 365805, 2009.
Article in English | MEDLINE | ID: mdl-20029641

ABSTRACT

A sixty-years-old male with diagnosis of a left adrenal mass (146 x 99 x 126 mm) with associated tumour thrombosis of the left renal vein with no clear signs of thrombosis of the inferior vena cava was admitted for elective surgery Finally an adrenalectomy and excision of tumour thrombus preserving the ipsilateral kidney was made. Despite of the complex vascular management, this kind of approaches allow to preserve normal renal function in patients with future nephrotoxic treatment like cisplatin.

19.
Rev. esp. patol ; 38(1): 48-50, ene.-mar. 2005. ilus
Article in Es | IBECS | ID: ibc-043954

ABSTRACT

Presentamos el caso de un varón de 70 años con un lipomade células fusiformes en la planta del pie, localizacióninfrecuente descrita hasta ahora tan sólo en tres ocasiones.Microscópicamente, el tumor está compuesto por tejidoadiposo maduro con células fusiformes acompañantes ymatriz mixoide rica en vasos. La inmunohistoquímicademuestra positividad de las células fusiformes para CD-34.El diagnóstico diferencial se plantea con el angiomixolipomay con tumores lipomatosos atípicos


We report the case of a 70-year-old man with a spindlecelllipoma on his right sole, a rare site that has heretoforebeen reported only in three instances. Microscopically, thetumor consists of mature adipose tissue with interspersedspindle cells, and features a myxoid matrix rich in bloodvessels. The immunohistochemical study reveals CD-34positivity in the spindle cells. The differential diagnosisincludes angiomyxolipoma and atypical lipomatoustumors


Subject(s)
Male , Aged , Humans , Lipoma/pathology , Nevus, Spindle Cell/pathology , Skin Neoplasms/pathology , Foot/pathology , Antigens, CD34/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...