Network


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

Hotspot


Dive into the research topics where Federico Pinna is active.

Publication


Featured researches published by Federico Pinna.


Gastroenterology | 2013

Yes-Associated Protein Up-regulates Jagged-1 and Activates the NOTCH Pathway in Human Hepatocellular Carcinoma

Darjus F. Tschaharganeh; Xin Chen; Philipp Latzko; Mona Malz; Matthias M. Gaida; Klaus Felix; Sara Ladu; Stephan Singer; Federico Pinna; Norbert Gretz; Carsten Sticht; Maria Lauda Tomasi; Salvatore Delogu; Matthias Evert; Biao Fan; Silvia Ribback; Lijie Jiang; Stefania Brozzetti; Frank Bergmann; Frank Dombrowski; Peter Schirmacher; Diego F. Calvisi; Kai Breuhahn

BACKGROUND & AIMS Cancer cells often lose contact inhibition to undergo anchorage-independent proliferation and become resistant to apoptosis by inactivating the Hippo signaling pathway, resulting in activation of the transcriptional co-activator yes-associated protein (YAP). However, the oncogenic mechanisms of YAP activity are unclear. METHODS By using cross-species analysis of expression data, the Notch ligand Jagged-1 (Jag-1) was identified as a downstream target of YAP in hepatocytes and hepatocellular carcinoma (HCC) cells. We analyzed the functions of YAP in HCC cells via overexpression and RNA silencing experiments. We used transgenic mice that overexpressed a constitutively activated form of YAP (YAP(S127A)), and measured protein levels in HCC, colorectal and pancreatic tumor samples from patients. RESULTS Human HCC cell lines and mouse hepatocytes that overexpress YAP(S127A) up-regulated Jag-1, leading to activation of the Notch pathway and increased proliferation. Induction of Jag-1, activation of Notch, and cell proliferation required binding of YAP to its transcriptional partner TEA domain family member 4 (TEAD4); TEAD4 binding required the Mst1/2 but not β-catenin signaling. Levels of YAP correlated with Jag-1 expression and Notch signaling in human tumor samples and correlated with shorter survival times of patients with HCC or colorectal cancer. CONCLUSIONS The transcriptional regulator YAP up-regulates Jag-1 to activate Notch signaling in HCC cells and mouse hepatocytes. YAP-dependent activity of Jag-1 and Notch correlate in human HCC and colorectal tumor samples with patient survival times, suggesting the use of YAP and Notch inhibitors as therapeutics for gastrointestinal cancer. Transcript profiling: microarray information was deposited at the Gene Expression Omnibus database (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?token=jxepvsumwosqkve&acc=GSE35004).


Developmental Biology | 2012

Insulin/IGF signaling drives cell proliferation in part via Yorkie/YAP

Katrin Straßburger; Marcel Tiebe; Federico Pinna; Kai Breuhahn; Aurelio A. Teleman

The insulin/IGF signaling (IIS) pathway is a potent inducer of cell proliferation in normal development and in cancer. The mechanism by which this occurs, however, is not completely understood. The Hippo signaling pathway regulates cell proliferation via the transcriptional co-activator Yorkie/YAP, however the signaling inputs regulating Hippo activity are not fully elucidated. Here we present evidence linking these two conserved, oncogenic pathways in Drosophila and in mammalian cells. We find that activation of IIS and of Yorkie signaling correlate positively in hepatocellular carcinoma. We show that IIS activates Yorkie in vivo, and that Yorkie plays an important role in the ability of IIS to drive cell proliferation. Interestingly, we also find the converse--that Yorkie signaling activates components of the insulin/TOR pathway. In sum, this crosstalk between IIS and Yorkie leads to coordinated regulation of these two oncogenic pathways.


Journal of Hepatology | 2015

Curcumin effectively inhibits oncogenic NF-κB signaling and restrains stemness features in liver cancer

Jens U. Marquardt; Luis Enrique Gómez-Quiroz; Lucrecia O. Arreguin Camacho; Federico Pinna; Yun-Han Lee; Mitsuteru Kitade; Mayrel Palestino Domínguez; D Castven; Kai Breuhahn; Elizabeth A. Conner; Peter R. Galle; Jesper B. Andersen; Valentina M. Factor; Snorri S. Thorgeirsson

BACKGROUND & AIMS The cancer stem cells (CSCs) have important therapeutic implications for multi-resistant cancers including hepatocellular carcinoma (HCC). Among the key pathways frequently activated in liver CSCs is NF-κB signaling. METHODS We evaluated the CSCs-depleting potential of NF-κB inhibition in liver cancer achieved by the IKK inhibitor curcumin, RNAi and specific peptide SN50. The effects on CSCs were assessed by analysis of side population (SP), sphere formation and tumorigenicity. Molecular changes were determined by RT-qPCR, global gene expression microarray, EMSA, and Western blotting. RESULTS HCC cell lines exposed to curcumin exhibited differential responses to curcumin and were classified as sensitive and resistant. In sensitive lines, curcumin-mediated induction of cell death was directly related to the extent of NF-κB inhibition. The treatment also led to a selective CSC-depletion as evidenced by a reduced SP size, decreased sphere formation, down-regulation of CSC markers and suppressed tumorigenicity. Similarly, NF-κB inhibition by SN50 and siRNA against p65 suppressed tumor cell growth. In contrast, curcumin-resistant cells displayed a paradoxical increase in proliferation and expression of CSC markers. Mechanistically, an important component of the CSC-depleting activity of curcumin could be attributed to a NF-κB-mediated HDAC inhibition. Co-administration of the class I/II HDAC inhibitor trichostatine sensitized resistant cells to curcumin. Further, integration of a predictive signature of curcumin sensitivity with human HCC database indicated that HCCs with poor prognosis and progenitor features are most likely to benefit from NF-κB inhibition. CONCLUSIONS These results demonstrate that blocking NF-κB can specifically target CSC populations and suggest a potential for combined inhibition of NF-κB and HDAC signaling for treatment of liver cancer patients with poor prognosis.


Hepatology | 2014

Prosurvival function of the cellular apoptosis susceptibility/importin‐α1 transport cycle is repressed by p53 in liver cancer

Juliane Winkler; Alessandro Ori; Kerstin Holzer; Carsten Sticht; Daniel Dauch; Eva Eiteneuer; Federico Pinna; Robert Geffers; Volker Ehemann; Amparo Andrés-Pons; Kai Breuhahn; Thomas Longerich; Justo Lorenzo Bermejo; Norbert Gretz; Lars Zender; Peter Schirmacher; Martin Beck; Stephan Singer

Proteins of the karyopherin superfamily including importins and exportins represent an essential part of the nucleocytoplasmic transport machinery. However, the functional relevance and regulation of karyopherins in hepatocellular carcinoma (HCC) is poorly understood. Here we identified cellular apoptosis susceptibility (CAS, exportin‐2) and its transport substrate importin‐α1 (imp‐α1) among significantly up‐regulated transport factor genes in HCC. Disruption of the CAS/imp‐α1 transport cycle by RNAi in HCC cell lines resulted in decreased tumor cell growth and increased apoptosis. The apoptotic phenotype upon CAS depletion could be recapitulated by direct knockdown of the X‐linked inhibitor of apoptosis (XIAP) and partially reverted by XIAP overexpression. In addition, XIAP and CAS mRNA expression levels were correlated in HCC patient samples (r = 0.463; P < 0.01), supporting the in vivo relevance of our findings. Furthermore, quantitative mass spectrometry analyses of murine HCC samples (p53−/− versus p53+/+) indicated higher protein expression of CAS and imp‐α1 in p53−/− tumors. Consistent with a role of p53 in regulating the CAS/imp‐α1 transport cycle, we observed that both transport factors were repressed upon p53 induction in a p21‐dependent manner. Conclusion: The CAS/imp‐α1 transport cycle is linked to XIAP and is required to maintain tumor cell survival in HCC. Moreover, CAS and imp‐α1 are targets of p53‐mediated repression, which represents a novel aspect of p53s ability to control tumor cell growth in hepatocarcinogenesis. (Hepatology 2014;60:884–895)


Journal of Hepatology | 2012

Transcriptional regulators in hepatocarcinogenesis--key integrators of malignant transformation.

Mona Malz; Federico Pinna; Peter Schirmacher; Kai Breuhahn

Hepatocellular carcinoma (HCC) is one of the most frequent human malignancies with poor prognosis and increasing incidence in the Western world. Only for a minority of HCC patients, surgical treatment options offer potential cure and therapeutic success of pharmacological approaches is limited. Highly specific approaches (e.g., kinase inhibitors) did not significantly improve the situation so far, possibly due to functional compensation, genetic heterogeneity of HCC, and development of resistance under selective pressure. In contrast, transcriptional regulators (especially transcription factors and co-factors) may integrate and process input signals of different (oncogenic) pathways and therefore represent cellular bottlenecks that regulate tumor cell biology. In this review, we want to summarize the current knowledge about central transcriptional regulators in human hepatocarcinogenesis and their potential as therapeutic target structures. Genomic and transcriptomic data of primary human HCC revealed that many of these factors showed up in subgroups of HCCs with a more aggressive phenotype, suggesting that aberrant activity of transcriptional regulators collect input information to promote tumor initiation and progression. Therefore, expression and dysfunction of transcription factors and co-factors may gain relevance for diagnostics and therapy of HCC.


Frontiers in Physiology | 2012

A Systems Biology Study on NFκB Signaling in Primary Mouse Hepatocytes.

Federico Pinna; Sven Sahle; Katharina Beuke; Michaela Bissinger; Selcan Tuncay; Lorenza A. D'Alessandro; Ralph Gauges; Andreas Raue; Jens Timmer; Ursula Klingmüller; Peter Schirmacher; Ursula Kummer; Kai Breuhahn

The cytokine tumor necrosis factor-alpha (TNFα) is one of the key factors during the priming phase of liver regeneration as well as in hepatocarcinogenesis. TNFα activates the nuclear factor κ-light-chain-enhancer of activated B cells (NFκB) signaling pathway and contributes to the conversion of quiescent hepatocytes to activated hepatocytes that are able to proliferate in response to growth factor stimulation. Different mathematical models have been previously established for TNFα/NFκB signaling in the context of tumor cells. Combining these mathematical models with time-resolved measurements of expression and phosphorylation of TNFα/NFκB pathway constituents in primary mouse hepatocytes revealed that an additional phosphorylation step of the NFκB isoform p65 has to be considered in the mathematical model in order to sufficiently describe the dynamics of pathway activation in the primary cells. Also, we addressed the role of basal protein turnover by experimentally measuring the degradation rate of pivotal players in the absence of TNFα and including this information in the model. To elucidate the impact of variations in the protein degradation rates on TNFα/NFκB signaling on the overall dynamic behavior we used global sensitivity analysis that accounts for parameter uncertainties and showed that degradation and translation of p65 had a major impact on the amplitude and the integral of p65 phosphorylation. Finally, our mathematical model of TNFα/NFκB signaling was able to predict the time-course of the complex formation of p65 and of the inhibitor of NFκB (IκB) in primary mouse hepatocytes, which was experimentally verified. Hence, we here present a mathematical model for TNFα/NFκB signaling in primary mouse hepatocytes that provides an important basis to quantitatively disentangle the complex interplay of multiple factors in liver regeneration and tumorigenesis.


PLOS ONE | 2014

Nuclear expression of the deubiquitinase CYLD is associated with improved survival in human hepatocellular carcinoma.

Stefan Ezechiel Welte; Toni Urbanik; Christin Elßner; Nicole Kautz; Bruno Christian Koehler; Nina Waldburger; Justo Lorenzo Bermejo; Federico Pinna; Karl-Heinz Weiss; Peter Schemmer; Dirk Jaeger; Thomas Longerich; Kai Breuhahn; Henning Schulze-Bergkamen

Background & Aims The deubiquitinase CYLD removes (K-63)-linked polyubiquitin chains from proteins involved in NF-κB, Wnt/ß-catenin and Bcl-3 signaling. Reduced CYLD expression has been reported in different tumor entities, including hepatocellular carcinoma (HCC). Furthermore, loss of CYLD has been shown to contribute to HCC development in knockout animal models. This study aimed to assess subcellular CYLD expression in tumor tissues and its prognostic significance in HCC patients undergoing liver resection or liver transplantation. Methods Subcellular localization of CYLD was assessed by immunohistochemistry in tumor tissues of 95 HCC patients undergoing liver resection or transplantation. Positive nuclear CYLD staining was defined as an immunhistochemical (IHC) score ≥3. Positive cytoplasmic CYLD staining was defined as an IHC score ≥6. The relationship with clinicopathological parameters was investigated. Cell culture experiments were performed to analyze subcellular CYLD expression in vitro. Results Cytoplasmic CYLD expression was observed in 57 out of 95 (60%) HCC specimens (cyt°CYLD+). Nuclear CYLD staining was positive in 52 out of 95 specimens (55%, nucCYLD+). 13 out of 52 nucCYLD+ patients (25%) showed a lack of cytoplasmic CYLD expression. nucCYLD+ was associated with prolonged overall survival in patients after resection or liver transplantation (P = 0.007). 5-year overall survival rates were 63% in nucCYLD+ vs. 26% in nucCYLD- patients. Nuclear CYLD staining strongly correlated with tumor grading (P<0.001) and Ki67 positivity (P = 0.005). nucCYLD+ did not prove to be an independent prognostic parameter. In vitro, Huh7, Hep3B and HepG2 showed reduced CYLD levels compared to the non-malignant liver cell line THLE-2. Induction of CYLD expression by doxorubicin treatment led to increased cytoplasmic and nuclear expression of CYLD. Conclusions Expression of nuclear CYLD is a novel prognostic factor for improved survival in patients with HCC undergoing liver resection or transplantation.


Hepatology | 2018

Cytoplasmic localization of the cell polarity factor Scribble supports liver tumor formation and tumor cell invasiveness

Shan Wan; Anne Sophie Meyer; S Weiler; Christian Rupp; Marcell Tóth; Carsten Sticht; Stephan Singer; Stefan Thomann; Stephanie Roessler; Marina Schorpp-Kistner; Jennifer Schmitt; Norbert Gretz; Peter Angel; Darjus F. Tschaharganeh; Jens U. Marquardt; Peter Schirmacher; Federico Pinna; Kai Breuhahn

The loss of epithelial cell polarity plays an important role in the development and progression of liver cancer. However, the specific molecular mechanisms supporting tumor initiation and progression are poorly understood. In this study, transcriptome data and immunofluorescence stains of tissue samples derived from hepatocellular carcinoma (HCC) patients revealed that overexpression associated with cytoplasmic localization of the basolateral cell polarity complex protein scribble (Scrib) correlated with poor prognosis of HCC patients. In comparison with HCC cells stably expressing wild‐type Scrib (ScribWT), mutated Scrib with enforced cytoplasmic enrichment (ScribP305L) induced AKT signaling through the destabilization of phosphatase and tensin homolog (PTEN) and PH domain and leucine‐rich repeat protein phosphatase 1 (PHLPP1). Cytoplasmic ScribP305L stimulated a gene signature and a phenotype characteristic for epithelial to mesenchymal transition (EMT) and HCC cell invasiveness. ScribP305L‐dependent invasion was mediated by the activator protein 1 (AP‐1) constituents ATF2 and JunB through induction of paracrine‐acting secreted protein acidic and cysteine‐rich (SPARC). Coexpression of ScribP305L and the oncogene c‐MYC through hydrodynamic gene delivery in mouse livers promoted tumor formation and increased abundance of pAKT, pATF2, and SPARC in comparison with controls. Finally, cytoplasmic Scrib localization correlated with AKT and ATF2 phosphorylation in human HCC tissues, and the ScribP305L‐dependent gene signature was enriched in cancer patients with poor prognosis. Conclusion: Perturbation of hepatocellular polarity due to overexpression and cytoplasmic enrichment of Scrib supports tumor initiation and HCC cell dissemination through specific molecular mechanisms. Biomarker signatures identified in this study can be used for the identification of HCC patients with higher risk for the development of metastasis. (Hepatology 2018;67:1842‐1856).


Gastroenterology | 2017

Induction of Chromosome Instability by Activation of Yes-Associated Protein and Forkhead Box M1 in Liver Cancer

S Weiler; Federico Pinna; Thomas Wolf; T Lutz; Aman Geldiyev; Carsten Sticht; M Knaub; Stefan Thomann; Michaela Bissinger; Shan Wan; Stephanie Rössler; D Becker; Norbert Gretz; Hauke Lang; Frank Bergmann; Vladimir Ustiyan; Tatiana V. Kalin; Stephan Singer; J. Lee; Jens U. Marquardt; Peter Schirmacher; Vladimir V. Kalinichenko; Kai Breuhahn


Cancer Research | 2013

Abstract 5235: A model for TNFα-mediated NFκB signalling: A systems biology study on hepatocytes and liver cancer cells.

Federico Pinna; Sven Sahle; Katharina Beuke; Michaela Bissinger; Selcan Tuncay; Lorenza A. D'Alessandro; Ralph Gauges; Andreas Raue; Jens Timmer; Ursula Klingmüller; Peter Schirmacher; Ursula Kummer; Kai Breuhahn

Collaboration


Dive into the Federico Pinna's collaboration.

Top Co-Authors

Avatar

Kai Breuhahn

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

Peter Schirmacher

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

S Weiler

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

Stephan Singer

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

T Lutz

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michaela Bissinger

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge