Network


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

Hotspot


Dive into the research topics where Giannicola Genovese is active.

Publication


Featured researches published by Giannicola Genovese.


Cell | 2013

The somatic genomic landscape of glioblastoma.

Cameron Brennan; Roel G.W. Verhaak; Aaron McKenna; Benito Campos; Houtan Noushmehr; Sofie R. Salama; Siyuan Zheng; Debyani Chakravarty; J. Zachary Sanborn; Samuel H. Berman; Rameen Beroukhim; Brady Bernard; Chang-Jiun Wu; Giannicola Genovese; Ilya Shmulevich; Jill S. Barnholtz-Sloan; Lihua Zou; Rahulsimham Vegesna; Sachet A. Shukla; Giovanni Ciriello; W.K. Yung; Wei Zhang; Carrie Sougnez; Tom Mikkelsen; Kenneth D. Aldape; Darell D. Bigner; Erwin G. Van Meir; Michael D. Prados; Andrew E. Sloan; Keith L. Black

We describe the landscape of somatic genomic alterations based on multidimensional and comprehensive characterization of more than 500 glioblastoma tumors (GBMs). We identify several novel mutated genes as well as complex rearrangements of signature receptors, including EGFR and PDGFRA. TERT promoter mutations are shown to correlate with elevated mRNA expression, supporting a role in telomerase reactivation. Correlative analyses confirm that the survival advantage of the proneural subtype is conferred by the G-CIMP phenotype, and MGMT DNA methylation may be a predictive biomarker for treatment response only in classical subtype GBM. Integrative analysis of genomic and proteomic profiles challenges the notion of therapeutic inhibition of a pathway as an alternative to inhibition of the target itself. These data will facilitate the discovery of therapeutic and diagnostic target candidates, the validation of research and clinical observations and the generation of unanticipated hypotheses that can advance our molecular understanding of this lethal cancer.


Nature | 2014

Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function

Andrea Viale; Piergiorgio Pettazzoni; Costas A. Lyssiotis; Haoqiang Ying; Nora Sanchez; Matteo Marchesini; Alessandro Carugo; Tessa Green; Sahil Seth; Virginia Giuliani; Maria Kost-Alimova; Florian Muller; Simona Colla; Luigi Nezi; Giannicola Genovese; Angela K. Deem; Avnish Kapoor; Wantong Yao; Emanuela Brunetto; Ya’an Kang; Min Yuan; John M. Asara; Y. Alan Wang; Timothy P. Heffernan; Alec C. Kimmelman; Huamin Wang; Jason B. Fleming; Lewis C. Cantley; Ronald A. DePinho; Giulio Draetta

Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers in western countries, with a median survival of 6 months and an extremely low percentage of long-term surviving patients. KRAS mutations are known to be a driver event of PDAC, but targeting mutant KRAS has proved challenging. Targeting oncogene-driven signalling pathways is a clinically validated approach for several devastating diseases. Still, despite marked tumour shrinkage, the frequency of relapse indicates that a fraction of tumour cells survives shut down of oncogenic signalling. Here we explore the role of mutant KRAS in PDAC maintenance using a recently developed inducible mouse model of mutated Kras (KrasG12D, herein KRas) in a p53LoxP/WT background. We demonstrate that a subpopulation of dormant tumour cells surviving oncogene ablation (surviving cells) and responsible for tumour relapse has features of cancer stem cells and relies on oxidative phosphorylation for survival. Transcriptomic and metabolic analyses of surviving cells reveal prominent expression of genes governing mitochondrial function, autophagy and lysosome activity, as well as a strong reliance on mitochondrial respiration and a decreased dependence on glycolysis for cellular energetics. Accordingly, surviving cells show high sensitivity to oxidative phosphorylation inhibitors, which can inhibit tumour recurrence. Our integrated analyses illuminate a therapeutic strategy of combined targeting of the KRAS pathway and mitochondrial respiration to manage pancreatic cancer.


Nature Medicine | 2012

Oncogenic NRAS signaling differentially regulates survival and proliferation in melanoma

Lawrence N. Kwong; James C. Costello; Huiyun Liu; Shan Jiang; Timothy L. Helms; Aliete E Langsdorf; David Jakubosky; Giannicola Genovese; Florian Muller; Joseph H. Jeong; Ryan P Bender; Gerald C. Chu; Keith T. Flaherty; Jennifer A. Wargo; James J. Collins; Lynda Chin

The discovery of potent inhibitors of the BRAF proto-oncogene has revolutionized therapy for melanoma harboring mutations in BRAF, yet NRAS-mutant melanoma remains without an effective therapy. Because direct pharmacological inhibition of the RAS proto-oncogene has thus far been unsuccessful, we explored systems biology approaches to identify synergistic drug combination(s) that can mimic RAS inhibition. Here, leveraging an inducible mouse model of NRAS-mutant melanoma, we show that pharmacological inhibition of mitogen-activated protein kinase kinase (MEK) activates apoptosis but not cell-cycle arrest, which is in contrast to complete genetic neuroblastoma RAS homolog (NRAS) extinction, which triggers both of these effects. Network modeling pinpointed cyclin-dependent kinase 4 (CDK4) as a key driver of this differential phenotype. Accordingly, combined pharmacological inhibition of MEK and CDK4 in vivo led to substantial synergy in therapeutic efficacy. We suggest a gradient model of oncogenic NRAS signaling in which the output is gated, resulting in the decoupling of discrete downstream biological phenotypes as a result of incomplete inhibition. Such a gated signaling model offers a new framework to identify nonobvious coextinction target(s) for combined pharmacological inhibition in NRAS-mutant melanomas.


Nature | 2012

Passenger deletions generate therapeutic vulnerabilities in cancer

Florian Muller; Simona Colla; Elisa Aquilanti; Veronica E. Manzo; Giannicola Genovese; Jaclyn Lee; Daniel Eisenson; Rujuta Narurkar; Pingna Deng; Luigi Nezi; Michelle Lee; Baoli Hu; Jian Hu; Ergun Sahin; Derrick Sek Tong Ong; Eliot Fletcher-Sananikone; Lawrence Kwong; Cameron Brennan; Y. Alan Wang; Lynda Chin; Ronald A. DePinho

Inactivation of tumour-suppressor genes by homozygous deletion is a prototypic event in the cancer genome, yet such deletions often encompass neighbouring genes. We propose that homozygous deletions in such passenger genes can expose cancer-specific therapeutic vulnerabilities when the collaterally deleted gene is a member of a functionally redundant family of genes carrying out an essential function. The glycolytic gene enolase 1 (ENO1) in the 1p36 locus is deleted in glioblastoma (GBM), which is tolerated by the expression of ENO2. Here we show that short-hairpin-RNA-mediated silencing of ENO2 selectively inhibits growth, survival and the tumorigenic potential of ENO1-deleted GBM cells, and that the enolase inhibitor phosphonoacetohydroxamate is selectively toxic to ENO1-deleted GBM cells relative to ENO1-intact GBM cells or normal astrocytes. The principle of collateral vulnerability should be applicable to other passenger-deleted genes encoding functionally redundant essential activities and provide an effective treatment strategy for cancers containing such genomic events.


Cancer Discovery | 2012

microRNA Regulatory Network Inference Identifies miR-34a as a Novel Regulator of TGF-β Signaling in Glioblastoma

Giannicola Genovese; Ayla Ergun; Sachet A. Shukla; Benito Campos; Jason A. Hanna; Papia Ghosh; Steven N. Quayle; Kunal Rai; Simona Colla; Haoquiang Ying; Chang-Jiun Wu; Sharmistha Sarkar; Yonghong Xiao; Jianhua Zhang; Hailei Zhang; Lawrence Kwong; Katherine Dunn; Wolf Ruprecht Wiedemeyer; Cameron Brennan; Hongwu Zheng; David L. Rimm; James J. Collins; Lynda Chin

UNLABELLED Leveraging The Cancer Genome Atlas (TCGA) multidimensional data in glioblastoma, we inferred the putative regulatory network between microRNA and mRNA using the Context Likelihood of Relatedness modeling algorithm. Interrogation of the network in context of defined molecular subtypes identified 8 microRNAs with a strong discriminatory potential between proneural and mesenchymal subtypes. Integrative in silico analyses, a functional genetic screen, and experimental validation identified miR-34a as a tumor suppressor in proneural subtype glioblastoma. Mechanistically, in addition to its direct regulation of platelet-derived growth factor receptor-alpha (PDGFRA), promoter enrichment analysis of context likelihood of relatedness-inferred mRNA nodes established miR-34a as a novel regulator of a SMAD4 transcriptional network. Clinically, miR-34a expression level is shown to be prognostic, where miR-34a low-expressing glioblastomas exhibited better overall survival. This work illustrates the potential of comprehensive multidimensional cancer genomic data combined with computational and experimental models in enabling mechanistic exploration of relationships among different genetic elements across the genome space in cancer. SIGNIFICANCE We illustrate here that network modeling of complex multidimensional cancer genomic data can generate a framework in which to explore the biology of cancers, leading to discovery of new pathogenetic insights as well as potential prognostic biomarkers. Specifically in glioblastoma, within the context of the global network, promoter enrichment analysis of network edges uncovered a novel regulation of TGF-β signaling via a Smad4 transcriptomic network by miR-34a.


Cell Reports | 2016

Post-translational Regulation of Cas9 during G1 Enhances Homology-Directed Repair

Tony Gutschner; Monika Haemmerle; Giannicola Genovese; Giulio Draetta; Lynda Chin

CRISPR/Cas9 induces DNA double-strand breaks that are repaired by cell-autonomous repair pathways, namely, non-homologous end-joining (NHEJ), or homology-directed repair (HDR). While HDR is absent in G1, NHEJ is active throughout the cell cycle and, thus, is largely favored over HDR. We devised a strategy to increase HDR by directly synchronizing the expression of Cas9 with cell-cycle progression. Fusion of Cas9 to the N-terminal region of human Geminin converted this gene-editing protein into a substrate for the E3 ubiquitin ligase complex APC/Cdh1, resulting in a cell-cycle-tailored expression with low levels in G1 but high expression in S/G2/M. Importantly, Cas9-hGem(1/110) increased the rate of HDR by up to 87% compared to wild-type Cas9. Future developments may enable high-resolution expression of genome engineering proteins, which might increase HDR rates further, and may contribute to a better understanding of DNA repair pathways due to spatiotemporal control of DNA damage induction.


Cancer Research | 2014

The RAC1 P29S Hotspot Mutation in Melanoma Confers Resistance to Pharmacological Inhibition of RAF

Ian R. Watson; Liren Li; Peter K. Cabeceiras; Mozhdeh Mahdavi; Tony Gutschner; Giannicola Genovese; Guocan Wang; Zhuangna Fang; James Tepper; Katherine Stemke-Hale; Kenneth Y. Tsai; Michael A. Davies; Gordon B. Mills; Lynda Chin

Following mutations in BRAF and NRAS, the RAC1 c.85C>T single-nucleotide variant (SNV) encoding P29S amino acid change represents the next most frequently observed protein-coding hotspot mutation in melanoma. However, the biologic and clinical significance of the RAC1 P29S somatic mutation in approximately 4% to 9% of patients remains unclear. Here, we demonstrate that melanoma cell lines possessing the RAC1 hotspot variant are resistant to RAF inhibitors (vemurafenib and dabrafenib). Enforced expression of RAC1 P29S in sensitive BRAF-mutant melanoma cell lines confers resistance manifested by increased viability, decreased apoptosis, and enhanced tumor growth in vivo upon treatment with RAF inhibitors. Conversely, RNAi-mediated silencing of endogenous RAC1 P29S in a melanoma cell line with a co-occurring BRAF V600 mutation increased sensitivity to vemurafenib and dabrafenib. Our results suggest RAC1 P29S status may offer a predictive biomarker for RAF inhibitor resistance in melanoma patients, where it should be evaluated clinically.


Cancer Cell | 2015

Telomere Dysfunction Drives Aberrant Hematopoietic Differentiation and Myelodysplastic Syndrome

Simona Colla; Derrick Sek Tong Ong; Yamini Ogoti; Matteo Marchesini; Nipun A. Mistry; Karen Clise-Dwyer; Sonny A. Ang; Paola Storti; Andrea Viale; Nicola Giuliani; Kathryn Ruisaard; Irene Ganan Gomez; Christopher A. Bristow; Marcos R. Estecio; David C. Weksberg; Yan Wing Ho; Baoli Hu; Giannicola Genovese; Piergiorgio Pettazzoni; Asha S. Multani; Shan Jiang; Sujun Hua; Michael C. Ryan; Alessandro Carugo; Luigi Nezi; Yue Wei; Hui Yang; Marianna D’Anca; Li Zhang; Sarah Gaddis

Myelodysplastic syndrome (MDS) risk correlates with advancing age, therapy-induced DNA damage, and/or shorter telomeres, but whether telomere erosion directly induces MDS is unknown. Here, we provide the genetic evidence that telomere dysfunction-induced DNA damage drives classical MDS phenotypes and alters common myeloid progenitor (CMP) differentiation by repressing the expression of mRNA splicing/processing genes, including SRSF2. RNA-seq analyses of telomere dysfunctional CMP identified aberrantly spliced transcripts linked to pathways relevant to MDS pathogenesis such as genome stability, DNA repair, chromatin remodeling, and histone modification, which are also enriched in mouse CMP haploinsufficient for SRSF2 and in CD34(+) CMML patient cells harboring SRSF2 mutation. Together, our studies establish an intimate link across telomere biology, aberrant RNA splicing, and myeloid progenitor differentiation.


Nature | 2017

Genomic deletion of malic enzyme 2 confers collateral lethality in pancreatic cancer

Prasenjit Dey; Joelle Baddour; Florian Muller; Chia Chin Wu; Huamin Wang; Wen Ting Liao; Zangdao Lan; Alina Chen; Tony Gutschner; Ya'an Kang; Jason B. Fleming; Nikunj Satani; Di Zhao; Abhinav Achreja; Lifeng Yang; Jiyoon Lee; Edward F. Chang; Giannicola Genovese; Andrea Viale; Haoqiang Ying; Giulio Draetta; Anirban Maitra; Y. Alan Wang; Deepak Nagrath; Ronald A. DePinho

The genome of pancreatic ductal adenocarcinoma (PDAC) frequently contains deletions of tumour suppressor gene loci, most notably SMAD4, which is homozygously deleted in nearly one-third of cases. As loss of neighbouring housekeeping genes can confer collateral lethality, we sought to determine whether loss of the metabolic gene malic enzyme 2 (ME2) in the SMAD4 locus would create cancer-specific metabolic vulnerability upon targeting of its paralogous isoform ME3. The mitochondrial malic enzymes (ME2 and ME3) are oxidative decarboxylases that catalyse the conversion of malate to pyruvate and are essential for NADPH regeneration and reactive oxygen species homeostasis. Here we show that ME3 depletion selectively kills ME2-null PDAC cells in a manner consistent with an essential function for ME3 in ME2-null cancer cells. Mechanistically, integrated metabolomic and molecular investigation of cells deficient in mitochondrial malic enzymes revealed diminished NADPH production and consequent high levels of reactive oxygen species. These changes activate AMP activated protein kinase (AMPK), which in turn directly suppresses sterol regulatory element-binding protein 1 (SREBP1)-directed transcription of its direct targets including the BCAT2 branched-chain amino acid transaminase 2) gene. BCAT2 catalyses the transfer of the amino group from branched-chain amino acids to α-ketoglutarate (α-KG) thereby regenerating glutamate, which functions in part to support de novo nucleotide synthesis. Thus, mitochondrial malic enzyme deficiency, which results in impaired NADPH production, provides a prime ‘collateral lethality’ therapeutic strategy for the treatment of a substantial fraction of patients diagnosed with this intractable disease.


Journal of Cellular Physiology | 2010

Post-translational modulation of CD133 expression during sodium butyrate-induced differentiation of HT29 human colon cancer cells: implications for its detection.

Alessandro Sgambato; Maria Ausiliatrice Puglisi; F Errico; Francesca Rafanelli; Alma Boninsegna; Alessandro Rettino; Giannicola Genovese; Claudio Coco; Antonio Gasbarrini; Achille Cittadini

The CD133 molecule has been proposed as a surface marker of cancer stem cells in several human malignancies, including colon cancers. The function and the mechanisms regulating CD133 expression remain unknown. The HT29 human colon cancer cells undergo differentiation following treatment with various agents and represent a useful in vitro model of colon differentiation. This study evaluated the behavior of CD133 during sodium butyrate‐induced differentiation of HT29 cells. Treatment with sodium butyrate induced a progressive decrease of CD133 expression, as assessed by flow cytometry using the AC133 monoclonal antibody. Indeed, expression of CD133, which was about 47% in untreated control cells, gradually decreased down to about 3% after 72 h in a time‐ and dose‐dependent manner. No relationship was observed between CD133 protein evaluated by flow cytometry and mRNA expression level, and no changes were detected in the methylation status of the CD133 gene promoter during HT29 differentiation. Moreover, the expression of the CD133 protein, evaluated by Western blot analysis using a specific anti‐CD133 antibody directed against the C‐terminal intracytoplasmic region of human CD133 protein, did not correlate with flow cytometry results. Different results were also obtained using the two antibodies to analyze the expression of the CD133 molecule in human colon cancers. These findings demonstrate that membrane expression of the CD133 stem cell marker might undergo a complex regulation during differentiation of colon cells and suggest that HT29 cells are a useful in vitro model to study the mechanisms involved in this regulation which likely occurs at a post‐transcriptional level. J. Cell. Physiol. 224:234–241, 2010

Collaboration


Dive into the Giannicola Genovese's collaboration.

Top Co-Authors

Avatar

Alessandro Carugo

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Giulio Draetta

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Lynda Chin

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sahil Seth

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Florian Muller

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Luigi Nezi

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Alessandro Sgambato

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar

Andrea Viale

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Simona Colla

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Cameron Brennan

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge