Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Alba Mota is active.

Publication


Featured researches published by Alba Mota.


The FASEB Journal | 2012

The protease MT1-MMP drives a combinatorial proteolytic program in activated endothelial cells

Agnieszka Koziol; Pilar Gonzalo; Alba Mota; Ángela Pollán; Cristina Lorenzo; Núria Colomé; David Montaner; Joaquín Dopazo; Joaquín Arribas; Francesc Canals; Alicia G. Arroyo

The mechanism by which proteolytic events translate into biological responses is not well understood. To explore the link of pericellular proteolysis to events relevant to capillary sprouting within the inflammatory context, we aimed at the identification of the collection of substrates of the protease MT1‐MMP in endothelial tip cells induced by inflammatory stimuli. We applied quantitative proteomics to endothelial cells (ECs) derived from wild‐type and MT1‐MMP‐null mice to identify the substrate repertoire of this protease in TNF‐α‐activated ECs. Bioinformatics analysis revealed a combinatorial MT1‐MMP proteolytic program, in which combined rather than single substrate processing would determine biological decisions by activated ECs, including chemotaxis, cell motility and adhesion, and vasculature development. MT1‐MMP‐deficient ECs inefficiently processed several of these substrates (TSP1, CYR61, NID1, and SEM3C), validating the model. This novel concept of MT1‐MMP‐driven combinatorial proteolysis in angiogenesis might be extendable to proteolytic actions in other cellular contexts.—Koziol, A., Gonzalo, P., Mota, A., Pollán, Á., Lorenzo, C., Colomé, N., Montaner, D., Dopazo, J., Arribas, J., Canals, F., Arroyo, A. G. The protease MT1‐MMP drives a combinatorial proteolytic program in activated endothelial cells. FASEB J. 26, 4481–4494 (2012). www.fasebj.org


PLOS ONE | 2014

Gasdermin-B Promotes Invasion and Metastasis in Breast Cancer Cells

Marta Hergueta-Redondo; David Sarrió; Ángela Molina-Crespo; Diego Megías; Alba Mota; Alejandro Rojo-Sebastián; Pablo García-Sanz; Saleta Morales; Sandra Abril; Amparo Cano; Héctor Peinado; Gema Moreno-Bueno

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.


EBioMedicine | 2016

Premalignant SOX2 overexpression in the fallopian tubes of ovarian cancer patients: Discovery and validation studies

Karin Hellner; Fabrizio Miranda; Donatien Fotso Chedom; Sandra Herrero-Gonzalez; Daniel M. Hayden; Rick Tearle; Mara Artibani; Mohammad KaramiNejadRanjbar; Ruth Williams; Kezia Gaitskell; Samar Elorbany; Ruoyan Xu; Alex Laios; Petronela Buiga; Karim Ahmed; Sunanda Dhar; Rebecca Yu Zhang; Leticia Campo; Kevin Alan Myers; Maria D. Lozano; María Ruiz-Miró; Sonia Gatius; Alba Mota; Gema Moreno-Bueno; Xavier Matias-Guiu; Javier Benitez; Lorna Witty; Gil McVean; Simon Leedham; Ian Tomlinson

Current screening methods for ovarian cancer can only detect advanced disease. Earlier detection has proved difficult because the molecular precursors involved in the natural history of the disease are unknown. To identify early driver mutations in ovarian cancer cells, we used dense whole genome sequencing of micrometastases and microscopic residual disease collected at three time points over three years from a single patient during treatment for high-grade serous ovarian cancer (HGSOC). The functional and clinical significance of the identified mutations was examined using a combination of population-based whole genome sequencing, targeted deep sequencing, multi-center analysis of protein expression, loss of function experiments in an in-vivo reporter assay and mammalian models, and gain of function experiments in primary cultured fallopian tube epithelial (FTE) cells. We identified frequent mutations involving a 40 kb distal repressor region for the key stem cell differentiation gene SOX2. In the apparently normal FTE, the region was also mutated. This was associated with a profound increase in SOX2 expression (p < 2−16), which was not found in patients without cancer (n = 108). Importantly, we show that SOX2 overexpression in FTE is nearly ubiquitous in patients with HGSOCs (n = 100), and common in BRCA1-BRCA2 mutation carriers (n = 71) who underwent prophylactic salpingo-oophorectomy. We propose that the finding of SOX2 overexpression in FTE could be exploited to develop biomarkers for detecting disease at a premalignant stage, which would reduce mortality from this devastating disease.


Modern Pathology | 2017

Genetic analysis of uterine aspirates improves the diagnostic value and captures the intra-tumor heterogeneity of endometrial cancers

Alba Mota; Eva Colas; Pablo García-Sanz; Irene Campoy; Alejandro Rojo-Sebastián; Sonia Gatius; Ángel García; Luis Chiva; Sonsoles Alonso; Xavier González-Tallada; Berta Díaz-Feijoo; August Vidal; Patrycja Ziober-Malinowska; Marcin Bobiński; Rafael Lopez-Lopez; Miguel Abal; Jaume Reventós; Xavier Matias-Guiu; Gema Moreno-Bueno

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.


Oncotarget | 2016

Gasdermin B expression predicts poor clinical outcome in HER2-positive breast cancer.

Marta Hergueta-Redondo; David Sarrió; Ángela Molina-Crespo; Rocio Vicario; Cristina Bernadó-Morales; Lidia Martínez; Alejandro Rojo-Sebastián; Jordi Serra-Musach; Alba Mota; Ángel Martínez-Ramírez; María Ángeles Castilla; Antonio Gonzalez-Martin; Sonia Pernas; Amparo Cano; Javier Cortes; Paolo Nuciforo; Vicente Peg; José Palacios; Miguel Angel Pujana; J. Arribas; Gema Moreno-Bueno

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.


Modern Pathology | 2015

A role for the transducer of the Hippo pathway, TAZ, in the development of aggressive types of endometrial cancer

Laura Romero-Pérez; Pablo García-Sanz; Alba Mota; Susanna Leskelä; Marta Hergueta-Redondo; Juan Díaz-Martín; M. Ángeles López-García; M Ángeles Castilla; Ángel Martínez-Ramírez; Robert A. Soslow; Xavier Matias-Guiu; Gema Moreno-Bueno; José Palacios

Although TAZ, the final effector of the Hippo pathway that modulates epithelial to mesenchymal transition and stemness, has been implicated in the development of different types of cancer, its role in endometrial cancer has not yet been studied. Thus, we evaluated the expression of TAZ in different types of endometrial cancer by immunohistochemistry. TAZ expression was detected in 76% of undifferentiated endometrial carcinomas, 54% of endometrial carcinosarcomas, 46% of endometrial serous carcinomas, 36% of grade 3 endometrioid carcinomas, and 18% of grade 1-2 endometrioid carcinomas, with statistically significant differences. We analyzed the WWTR1 gene that encodes TAZ by FISH and MassARRAY spectrometry, ruling out gene amplification and differential promoter methylation as the main mechanisms that modulate TAZ expression in endometrial tumors. However, we did detect a significant association between Scribble hypoexpression and delocalization with TAZ expression. Moreover, we demonstrated that TAZ promoted invasiveness, and it favored cell motility and tumor growth, in endometrial cancer cell lines. In addition, TAZ expression was associated with the transition from an epithelial to mesenchymal phenotype, both in vitro and in human tumors. Together, these data reveal a previously unknown role for TAZ and the Hippo pathway in the progression of aggressive subtypes of endometrial cancer.


Human Pathology | 2014

A 9-protein biomarker molecular signature for predicting histologic type in endometrial carcinoma by immunohistochemistry.

Maria Santacana; Oscar Maiques; Joan Valls; Sonia Gatius; Ana Isabel Abó; María Ángeles López-García; Alba Mota; Jaume Reventós; Gema Moreno-Bueno; José Palacios; Carla Bartosch; Xavier Dolcet; Xavier Matias-Guiu

Histologic typing may be difficult in a subset of endometrial carcinoma (EC) cases. In these cases, interobserver agreement improves when immunohistochemistry (IHC) is used. A series of endometrioid type (EEC) grades 1, 2, and 3 and serous type (SC) were immunostained for p53, p16, estrogen receptor, PTEN, IMP2, IMP3, HER2, cyclin B2 and E1, HMGA2, FolR1, MSLN, Claudins 3 and 4, and NRF2. Nine biomarkers showed significant differences with thresholds in IHC value scale between both types (p53 ≥ 20, IMP2 ≥ 115, IMP3 ≥ 2, cyclin E1 ≥ 220, HMGA2 ≥ 30, FolR1 ≥ 50, p16 ≥ 170, nuclear PTEN ≥ 2 and estrogen receptor ≤ 50; P < .005). This combination led to increased discrimination when considering cases satisfying 0 to 5 conditions predicted as EEC and those satisfying 6 to 9 conditions predicted as SC. This signature correctly predicted all 48 EEC grade 1-2 cases and 18 SC cases, but 3 SC cases were wrongly predicted as EEC. Sensitivity was 86% (95% confidence interval [CI], 64%-97%), and specificity was 100% (95% CI, 89%-100%). The classifier correctly predicted all 28 EEC grade 3 cases but only identified the EEC and SC components in 4 of 9 mixed EEC-SC. An independent validation series (29 EEC grades 1-2, 28 EEC grade 3, and 31 SC) showed 100% sensitivity (95% CI, 84%-100%) and 83% specificity (95% CI, 64%-94%). We propose an internally and externally validated 9-protein biomarker signature to predict the histologic type of EC (EEC or SC) by IHC. The results also suggest that mixed EEC-SC is molecularly ambiguous.


BMC Cancer | 2015

Intra-tumor heterogeneity in TP53 null High Grade Serous Ovarian Carcinoma progression

Alba Mota; Juan Carlos Triviño; Alejandro Rojo-Sebastián; Ángel Martínez-Ramírez; Luis Chiva; Antonio Gonzalez-Martin; Juan F. García; Pablo García-Sanz; Gema Moreno-Bueno

BackgroundHigh 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.MethodsIn 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.ResultsSignificant 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.ConclusionsAltogether, 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.


International Journal of Cancer | 2017

Chromatin remodelling and DNA repair genes are frequently mutated in endometrioid endometrial carcinoma.

Pablo García-Sanz; Juan Carlos Triviño; Alba Mota; María Pérez López; Eva Colas; Alejandro Rojo-Sebastián; Angel Garcia; Sonia Gatius; Maria Cristina Ruiz; Jaime Prat; Rafael Lopez-Lopez; Miguel Abal; Jaume Reventós; Xavier Matias-Guiu; Gema Moreno-Bueno

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.


International Journal of Gynecological Cancer | 2016

Biological Effects of Temsirolimus on the mTOR Pathway in Endometrial Carcinoma: A Pharmacodynamic Phase II Study.

Maria Santacana; Pluvio J. Coronado; Xavier Matias-Guiu; Ignacio Romero; Antonio Casado; Ma Alba Dosil; Alba Mota; Gema Moreno-Bueno; Xavier Dolcet; Antonio Llombart-Cussac; Andres Poveda

OBJECTIVE The PI3K/AKT/mTOR pathway is frequently aberrantly activated in endometrial carcinoma (EC). Temsirolimus is an mTOR inhibitor that has shown clinical activity in EC. We aimed to characterize the biological effects on mTOR pathway of temsirolimus in treatment-naive patients with primary EC, and to identify potential biomarkers associated with a short-term exposure to temsirolimus. MATERIALS AND METHODS Patients with EC were treated with 4 doses of temsirolimus previous to surgery. The primary objective was the analysis of paired endometrial aspirates and posttreatment (hysterectomy specimens) tumor tissue samples for mTOR downstream effectors p-S6K1 and p-4BEP1 levels by immunohistochemistry. Secondary objectives included analysis of expression of other mTOR-related biomarkers by immunohistochemistry, as well as analysis of the predictive value of mutations in mTOR-related genes. Toxicity was also assessed. RESULTS Eleven patients were included in the study. p-S6K1 expression was reduced after treatment with temsirolimus in all patients. Variations of the expression of other mTOR-related proteins including p-4BEP1, PTEN, p-AKT, p53, p27, BAD, Bcl-2, Ki67, and cyclin D1 were also observed. Interestingly, the biological effects of the drug were more evident 1 week after the last dose of temsirolimus. Effects were less evident on tumors harboring mutations in NRAS. Toxicity was acceptable, being mucositis the most frequent adverse event. CONCLUSIONS Short temsirolimus exposure effectively inhibits mTOR pathway in patients with endometrial cancer. p-S6K1 expression is a promising biomarker of sensitivity. The preoperative window opportunity in EC is a realistic scenario for biological knowledge and target development.

Collaboration


Dive into the Alba Mota's collaboration.

Top Co-Authors

Avatar

Gema Moreno-Bueno

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Alejandro Rojo-Sebastián

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Xavier Matias-Guiu

Hospital Universitari Arnau de Vilanova

View shared research outputs
Top Co-Authors

Avatar

Pablo García-Sanz

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Marta Hergueta-Redondo

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Sonia Gatius

Hospital Universitari Arnau de Vilanova

View shared research outputs
Top Co-Authors

Avatar

Amparo Cano

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Jaume Reventós

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ángela Molina-Crespo

Spanish National Research Council

View shared research outputs
Researchain Logo
Decentralizing Knowledge