Network


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

Hotspot


Dive into the research topics where David Llobet is active.

Publication


Featured researches published by David Llobet.


Virchows Archiv | 2005

NF-kB in development and progression of human cancer

Xavier Dolcet; David Llobet; Judit Pallares; Xavier Matias-Guiu

The nuclear factor kB (NF-kB) comprises a family of transcription factors involved in the regulation of a wide variety of biological responses. NF-kB plays a well-known function in the regulation of immune responses and inflammation, but growing evidences support a major role in oncogenesis. NF-kB regulates the expression of genes involved in many processes that play a key role in the development and progression of cancer such as proliferation, migration and apoptosis. Aberrant or constitutive NF-kB activation has been detected in many human malignancies. In recent years, numerous studies have focused on elucidating the functional consequences of NF-kB activation as well as its signaling mechanisms. NF-kB has turned out to be an interesting therapeutic target for treatment of cancer.


Journal of Clinical Pathology | 2009

Molecular pathology of endometrial carcinoma: practical aspects from the diagnostic and therapeutic viewpoints

David Llobet; Judith Pallares; Maria Santacana; Nuria Eritja; Ana Velasco; Xavier Dolcet; Xavier Matias-Guiu

This article reviews the main molecular alterations involved in endometrial carcinoma. Five molecular features (microsatellite instability, and mutations in the PTEN, k-RAS, PIK3CA and β-catenin genes) are characteristic of endometrioid carcinomas, whereas non-endometrioid carcinomas show alterations of p53, loss of heterozygosity (LOH) on several chromosomes, as well as other molecular alterations (STK15, p16, E-cadherin and C-erb B2). The review also covers the phenomenon of apoptosis resistance, as well as the results obtained from cDNA array studies, and the perspectives for targeted therapies. A group of practical applications of molecular pathology techniques are also mentioned: diagnosis of hereditary non-polyposis colon cancer syndrome in patients with endometrial carcinoma; evaluation of precursor lesions; prognosis; diagnosis, particularly for synchronous endometrioid carcinomas of the uterus and the ovaries; and targeted therapies.


Journal of Biological Chemistry | 2006

Proteasome Inhibitors Induce Death but Activate NF-κB on Endometrial Carcinoma Cell Lines and Primary Culture Explants

Xavier Dolcet; David Llobet; Mario Encinas; Judit Pallares; Albert Cabero; Joan Antoni Schoenenberger; Joan X. Comella; Xavier Matias-Guiu

Proteasome inhibitors are currently used as chemotherapeutic drugs because of their ability to block NF-κB, a transcription factor constitutively activated in many different types of human cancer. In the present study, we demonstrate that proteasome inhibitors induce cell death in endometrial carcinoma cell lines and primary explants but, instead of blocking NF-κB, they increase its transcriptional activity. Proteasome inhibitors induce phosphorylation of IKKα/β, phosphorylation and degradation of IκBα, and phosphorylation of the p65 NF-κB subunit on serine 536. Proteasome inhibitor-induced NF-κB activity can be blocked by a non-degradable form of IκBα or dominant negative forms of either IKKα or IKKβ. Lentiviral delivery of shRNAs to either IKKα or IKKβ cause blockade of NF-κB transcriptional activity and inhibit phosphorylation of p65 on serine 536, but has no effect on IκBa degradation. These results suggest a role for p65 phosphorylation in proteasome inhibitor-induced NF-κB activation. Accordingly, siRNA knockdown of p65 inhibits proteasome inhibitor-induced NF-κB transcriptional activity. Our results demonstrate that proteasome inhibitors, including bortezomib, induce cell death on endometrial carcinoma cells and primary explants. However, they activate NF-κB instead of blocking its transcriptional potential. Therefore, the concept that proteasome inhibitors are blockers of NF-κB activation should be carefully examined in particular cell types.


Modern Pathology | 2005

Immunohistochemical analysis of PTEN in endometrial carcinoma: a tissue microarray study with a comparison of four commercial antibodies in correlation with molecular abnormalities

Judit Pallares; Elena Bussaglia; Jose Luis Martínez-Guitarte; Xavier Dolcet; David Llobet; Montserrat Rue; Lidia Sanchez-Verde; José Palacios; Jaime Prat; Xavier Matias-Guiu

The tumor suppressor gene PTEN/MMAC1 is located on chromosome 10q23.3. Inactivation of PTEN, either by mutations, deletions, or promoter hypermethylation, has been identified in a wide variety of tumors. Inactivation of the two alleles of PTEN is required, because it is a tumor suppressor gene. Immunohistochemical staining may be an effective screening method to demonstrate the absence of the protein in tumors exhibiting PTEN inactivation. We studied a tissue microarray, constructed from paraffin-embedded blocks of 95 endometrial carcinomas, 38 of them previously evaluated for alterations in PTEN. We also studied cell blocks obtained from one PTEN-defective endometrial cancer cell line, after transfection with either a plasmid encoding wild-type PTEN or the empty vector. The tumor samples were tested with four different anti-PTEN commercial antibodies: a polyclonal antibody, the monoclonal antibody 28H6, the monoclonal antibody 10P03, and the monoclonal antibody 6.H2.1. Results were correlated with the presence of abnormalities in PTEN, as well as with the immunohistochemical expression of phosphorylated AKT. Antibody 28H6 produced a predominant nuclear staining, while the other three antibodies produced a predominant cytoplasmic staining. There was no significant correlation between the results obtained with the four antibodies. The monoclonal antibody 6.H2.1 was the only one that exhibited a correlation with the presence of molecular alterations in PTEN, and a statistically significant association with immunostaining for phosphorylated AKT (r=−0.249, P=0.037). The monoclonal antibody 10P03 exhibited an association with phospho-AKT that did not have statistical significance. Both 6.H2.1 and 10P03 antibodies stained PTEN-transfected cells, and were negative in the PTEN-deficient cell line blocks. The polyclonal antibody and the monoclonal antibody 28H6 produced positive staining in PTEN-deficient cell line blocks, suggesting nonspecific staining. The results indicate that monoclonal antibody 6.H2.1 may be a suitable alternative for tumors with inactivation of PTEN.


The Journal of Pathology | 2004

Abnormalities in the NF-κB family and related proteins in endometrial carcinoma

Judit Pallares; Jose Luis Martínez-Guitarte; Xavier Dolcet; David Llobet; Montserrat Rue; José Palacios; Jaime Prat; Xavier Matias-Guiu

The NF‐κB family of transcription factors regulates a wide variety of cellular processes including cell growth, differentiation, and apoptosis. A tissue microarray was constructed from paraffin wax‐embedded blocks from 95 endometrial carcinomas (EC), previously studied for microsatellite instability, as well as for alterations in PTEN, k‐RAS and beta‐catenin. Immunohistochemical evaluation included members of the NF‐κB (p50, p65, p52, c‐Rel, Rel‐B) and the IκB (IκBα, IκBβ, IκBε, Bcl‐3) families, as well as putative targets of NF‐κB such as Flip, Bcl‐xL, Cyclin D1, and oestrogen and progesterone receptors. Results were correlated with the clinical and pathological data. Nuclear immunostaining for members of the NF‐κB family was frequent in EC (p50, 20%; p65, 16.5–21.9%; p52, 9.3%; c‐Rel, 48.9%; Rel‐B, 15.7%); and it correlated with negativity for members of the IκB family in some cases. There was a statistically significant association between immunoreaction for p50 and p65 (p = 0.006), suggesting activation of the so‐called ‘classic form’ of NF‐κB, similar to that described in breast cancer. Bcl‐3 nuclear immunostaining was detected in 60.7% of cases. The vast majority of p52‐positive tumours showed Bcl‐3 nuclear immunoreaction (p = 0.038). Immunostaining for putative targets of NF‐κB was as follows: Bcl‐xL, 76.2% (p = 0.001); Flip 43.0%; Cyclin D1, 64.79%. p65 immunostaining correlated with increased immunoreaction for steroid hormone receptors. No correlation was found between NF‐κB nuclear pattern and the presence of microsatellite instability, or alterations in PTEN, k‐RAS, or beta‐catenin. These results suggest that the NF‐κB and IκB families of genes may be important in endometrial carcinogenesis, by controlling apoptosis and cell proliferation. Copyright


Laboratory Investigation | 2005

FLIP is frequently expressed in endometrial carcinoma and has a role in resistance to TRAIL-induced apoptosis

Xavier Dolcet; David Llobet; Judit Pallares; Montserrat Rue; Joan X. Comella; Xavier Matias-Guiu

The FLICE-inhibitory protein (FLIP) plays a key role in the regulation of apoptosis triggered by death ligands. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been shown to induce apoptosis in some types of tumor but not in others. To assess the possible role of FLIP in apoptosis resistance in endometrial carcinoma, we performed an immunohistochemical study on a tissue microarray composed of 95 endometrial carcinomas. We found positive signals in 43% of the cases, as well as a significant difference in FLIP expression between stage I and II tumors. Moreover, we observed that endometrial carcinoma cell lines Ishikawa and KLE did not undergo apoptosis after TRAIL treatment. Cotreatment of these cells with the inhibitor of transcription actinomycin D resulted in a dramatic decrease in cell viability and induced activation of caspase-8. These events coincided with downregulation of FLIP mRNA and protein. Inhibitors of caspase-8 or overexpression of FLIP completely blocked apoptosis induced by actinomycin D plus TRAIL cotreatment. More importantly, downregulation of endogenous FLIP expression by specific siRNAs sensitized endometrial carcinoma cells to TRAIL-induced apoptosis in the absence of actinomycin D. Taken together, our results suggest for the first time a critical role for FLIP in the regulation apoptosis triggered by TRAIL in endometrial carcinoma cells.


International Journal of Gynecological Pathology | 2005

Survivin expression in endometrial carcinoma: a tissue microarray study with correlation with PTEN and STAT-3.

Judit Pallares; Jose Luis Martínez-Guitarte; Xavier Dolcet; David Llobet; Montserrat Rue; José Palacios; Jaime Prat; Xavier Matias-Guiu

Evasion of apoptotic cell death plays a key role in cancer development. Survivin is a member of the inhibitor of apoptosis proteins, which also has a role in the control of cell division. Survivin may be overexpressed in some tumors and has been suggested to be related to PTEN, β-catenin, p53 [all of them frequently abnormal in endometrial carcinomas (ECs)], and STAT-3. A tissue microarray was constructed from paraffin-embedded blocks of 95 ECs, previously studied for microsatellite instability and for alterations in PTEN, k-RAS, and CTNNB-1. Immunohistochemical evaluation included 1) survivin, 2) markers of cell proliferation and apoptosis (Ki67-MIB1 and M 30-neoepitope cytokeratin 18), and 3) proteins involved in cell signaling pathways (PTEN, phospho-AKT, β-catenin, p53, and STAT-3). Survivin expression was frequent in ECs (75.95%) but did not show any statistical significant correlation with histological type and grade, stage, overall survival, or mitotic and apoptotic indexes. Survivin expression had a statistical significant correlation with decreased PTEN expression (r = −0.383, p = 0.001), increased phospho-AKT (r = 0.70, p < 0.001), and positive STAT-3 immunostaining (r = 0.6, p < 0.001). Survivin expression did not show statistical correlation with either β-catenin or p53 alterations. The results suggest that increased survivin expression is frequent in ECs and may be dependent on STAT-3 and PI3 K/AKT activation. Because PTEN abnormalities are very frequent in ECs, the results from this study indicate that PTEN may interfere with the process of apoptosis and cell proliferation by promoting survivin expression.


Human Pathology | 2012

Epithelial to mesenchymal transition in early stage endometrioid endometrial carcinoma

Núria Montserrat; Ana Mozos; David Llobet; Xavier Dolcet; Cristina Pons; Antonio García de Herreros; Xavier Matias-Guiu; Jaime Prat

Epithelial to mesenchymal transition is thought to be implicated in tumor invasion and metastasis. To investigate its role in myometrial invasion, samples from 42 stage I (confined to the corpus) endometrioid endometrial carcinomas were analyzed. All E-cadherin repressors (SNAI1, SNAI2 (SLUG), ZEB1, HMGA2, and TWIST1) had a higher expression in endometrioid endometrial carcinomas than in normal endometrium (P < .0001), whereas CDH1 (E-cadherin gene) tended to be lower. In comparison with nonmyoinvasive (stage IA) tumors, those with deep myometrial invasion (stage IC) had increased messenger RNA expression of SLUG, ZEB1, and HMGA2 (P < .001). Furthermore, samples from the myoinvasive front of deeply invasive tumors had higher levels of SLUG, ZEB1, and HMGA2 than the corresponding superficial samples. Immunohistochemical analysis of these cases revealed that the decrease in E-cadherin was concordant with an increase in Snail and Twist protein expression. Trying to induce epithelial to mesenchymal transition in endometrioid endometrial carcinomas, we initially produced persistent activation of this pathway in Ishikawa cells. The cell line was infected with lentiviruses carrying the V600E mutation of BRAF, inducing loss of β-catenin, E-cadherin, and cytokeratin and increase in vimentin and Snail. These changes were mediated by ERK1/2 phosphorylation, which was also increased at the myoinvasive front. Furthermore, MEK1/2 inhibitor UO126 reversed the mesenchymal phenotype. Our findings suggest that epithelial to mesenchymal transition regulators are implicated in myometrial invasion of endometrioid endometrial carcinoma and may be potential therapeutic targets through the MAPK/ERK pathway.


Oncogene | 2008

CK2 controls TRAIL and Fas sensitivity by regulating FLIP levels in endometrial carcinoma cells.

David Llobet; Nuria Eritja; Mario Encinas; Nuria Llecha; Judit Pallares; Anabel Sorolla; F.J. Gonzalez-Tallada; Xavier Matias-Guiu; Xavier Dolcet

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has emerged as a promising antineoplastic agent because of its ability to selectively kill tumoral cells. However, some cancer cells are resistant to TRAIL-induced apoptosis. We have previously demonstrated that in endometrial carcinoma cells such resistance is caused by elevated FLICE-inhibitory protein (FLIP) levels. The present study focuses on the mechanisms by which FLIP could be modulated to sensitize endometrial carcinoma cells to TRAIL-induced apoptosis. We find that inhibition of casein kinase (CK2) sensitizes endometrial carcinoma cells to TRAIL- and Fas-induced apoptosis. CK2 inhibition correlates with a reduction of FLIP protein, suggesting that CK2 regulates resistance to TRAIL and Fas by controlling FLIP levels. FLIP downregulation correlates with a reduction of mRNA and is prevented by addition of the MG-132, suggesting that CK2 inhibition results in a proteasome-mediated degradation of FLIP. Consistently, forced expression of FLIP restores resistance to TRAIL and Fas. Moreover, knockdown of either FADD or caspase-8 abrogates apoptosis triggered by inhibition of CK2, indicating that CK2 sensitization requires formation of functional DISC. Finally, because of the possible role of both TRAIL and CK2 in cancer therapy, we demonstrate that CK2 inhibition sensitizes primary endometrial carcinoma explants to TRAIL apoptosis. In conclusion, we demonstrate that CK2 regulates endometrial carcinoma cell sensitivity to TRAIL and Fas by regulating FLIP levels.


Anti-Cancer Drugs | 2008

Antioxidants block proteasome inhibitor function in endometrial carcinoma cells

David Llobet; Nuria Eritja; Mario Encinas; Anabel Sorolla; Joan Antoni Schoenenberger; Antonio Llombart-Cussac; Rosa M. Martí; Xavier Matias-Guiu; Xavier Dolcet

We have recently demonstrated that proteasome inhibitors can be effective in inducing apoptotic cell death in endometrial carcinoma cell lines and primary culture explants. Increasing evidence suggests that reactive oxygen species are responsible for proteasome inhibitor-induced cell killing. Antioxidants can thus block apoptosis (cell death) triggered by proteasome inhibition. Here, we have evaluated the effects of different antioxidants (edaravone and tiron) on endometrial carcinoma cells treated with aldehyde proteasome inhibitors (MG-132 or ALLN), the boronic acid-based proteasome inhibitor (bortezomib) and the epoxyketone, epoxomicin. We show that tiron specifically inhibited the cytotoxic effects of bortezomib, whereas edaravone inhibited cell death caused by aldehyde-based proteasome inhibitors. We have, however, found that edaravone completely inhibited accumulation of ubiquitin and proteasome activity decrease caused by MG-132 or ALLN, but not by bortezomib. Conversely, tiron inhibited the ubiquitin accumulation and proteasome activity decrease caused by bortezomib. These results suggest that edaravone and tiron rescue cells of proteasome inhibitors from cell death, by inhibiting blockade of proteasome caused by MG-132 and ALLN or bortezomib, respectively. We also tested other antioxidants, and we found that vitamin C inhibited bortezomib-induced cell death. Similar to tiron, vitamin C inhibited cell death by blocking the ability of bortezomib to inhibit the proteasome. Until now, all the antioxidants that blocked proteasome inhibitor-induced cell death also blocked the proteasome inhibitor mechanism of action.

Collaboration


Dive into the David Llobet's collaboration.

Top Co-Authors

Avatar

Xavier Matias-Guiu

Hospital Universitari Arnau de Vilanova

View shared research outputs
Top Co-Authors

Avatar

Xavier Dolcet

Hospital Universitari Arnau de Vilanova

View shared research outputs
Top Co-Authors

Avatar

Judit Pallares

Hospital Universitari Arnau de Vilanova

View shared research outputs
Top Co-Authors

Avatar

Nuria Eritja

Hospital Universitari Arnau de Vilanova

View shared research outputs
Top Co-Authors

Avatar

Maria Santacana

Hospital Universitari Arnau de Vilanova

View shared research outputs
Top Co-Authors

Avatar

Anabel Sorolla

Hospital Universitari Arnau de Vilanova

View shared research outputs
Top Co-Authors

Avatar

Mario Encinas

Hospital Universitari Arnau de Vilanova

View shared research outputs
Top Co-Authors

Avatar

Jaime Prat

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mónica Domingo

Hospital Universitari Arnau de Vilanova

View shared research outputs
Researchain Logo
Decentralizing Knowledge