40 results on '"Carla Danussi"'
Search Results
2. Atrx inactivation drives disease-defining phenotypes in glioma cells of origin through global epigenomic remodeling
- Author
-
Carla Danussi, Promita Bose, Prasanna T. Parthasarathy, Pedro C. Silberman, John S. Van Arnam, Mark Vitucci, Oliver Y. Tang, Adriana Heguy, Yuxiang Wang, Timothy A. Chan, Gregory J. Riggins, Erik P. Sulman, Frederick Lang, Chad J. Creighton, Benjamin Deneen, C. Ryan Miller, David J. Picketts, Kasthuri Kannan, and Jason T. Huse
- Subjects
Science - Abstract
ATRX inactivation frequently occurs in glioma. Here, the authors explore the role of ATRX inactivation in oncogenesis, highlighting ATRX deficiency driven epigenomic changes that influence the expression of genes crucial to the oncogenic phenotype.
- Published
- 2018
- Full Text
- View/download PDF
3. Identification of patient-derived glioblastoma stem cell (GSC) lines with the alternative lengthening of telomeres phenotype
- Author
-
Ahsan Farooqi, Jie Yang, Vladislav Sharin, Ravesanker Ezhilarasan, Carla Danussi, Christian Alvarez, Sharvari Dharmaiah, David Irvin, Jason Huse, and Erik P. Sulman
- Subjects
Neurology. Diseases of the nervous system ,RC346-429 - Published
- 2019
- Full Text
- View/download PDF
4. Author Correction: Atrx inactivation drives disease-defining phenotypes in glioma cells of origin through global epigenomic remodeling
- Author
-
Carla Danussi, Promita Bose, Prasanna T. Parthasarathy, Pedro C. Silberman, John S. Van Arnam, Mark Vitucci, Oliver Y. Tang, Adriana Heguy, Yuxiang Wang, Timothy A. Chan, Gregory J. Riggins, Erik P. Sulman, Frederick F. Lang, Chad J. Creighton, Benjamin Deneen, C. Ryan Miller, David J. Picketts, Kasthuri Kannan, and Jason T. Huse
- Subjects
Science - Published
- 2022
- Full Text
- View/download PDF
5. Novel insights into the epigenetics of diffuse glioma
- Author
-
Carla Danussi and Jason T. Huse
- Subjects
atrx ,glioma ,cancer genomics ,epigenomics ,histones ,neuroepithelial progenitors ,migration ,differentiation ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Loss-of-function mutations of the chromatin regulator ATRX (α-thalassemia mental retardation X-linked) occur frequently in diffuse gliomas, but the molecular mechanisms by which ATRX inactivation promotes oncogenesis remain unclear. We recently reported that Atrx deficiency drives glioma-relevant phenotypes, such as increased motility and astrocytic differentiation profiles, by directly modulating epigenomic lanscapes in glioma cells of origin. Our work has significant implications on the role of epigenetic regulator dysfunction in the oncogenic process.
- Published
- 2018
- Full Text
- View/download PDF
6. Supplementary Figures 1-3 from An EMILIN1-Negative Microenvironment Promotes Tumor Cell Proliferation and Lymph Node Invasion
- Author
-
Paola Spessotto, Alfonso Colombatti, Lisa Del Bel Belluz, Eliana Pivetta, Teresa Maria Elisa Modica, Bruna Wassermann, Alessandra Petrucco, and Carla Danussi
- Abstract
PDF file - 274K, Additional information for Figure 3 and 6 and full length blots for Figure 2K and Figure 4C
- Published
- 2023
- Full Text
- View/download PDF
7. Supplemental Methods and References,Supplementary Figures S1-S3, and Supplementary Table S1 from RHPN2 Drives Mesenchymal Transformation in Malignant Glioma by Triggering RhoA Activation
- Author
-
Antonio Iavarone, Dana Pe'er, Anna Lasorella, Andreja Jovic, Francesco Niola, Uri David Akavia, and Carla Danussi
- Abstract
Supplemental Methods and References. Figure S1. Illustration of Module generation Figure S2. CNV and Expression of RHPN2 in GBM samples from TCGA and Rembrandt databases. Figure S3. Western Blot and qPCR of RHPN2 in different cell lines. Table S1. List of Primers
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Table S2 from RHPN2 Drives Mesenchymal Transformation in Malignant Glioma by Triggering RhoA Activation
- Author
-
Antonio Iavarone, Dana Pe'er, Anna Lasorella, Andreja Jovic, Francesco Niola, Uri David Akavia, and Carla Danussi
- Abstract
Table S2. Drivers identified by Multi-Reg
- Published
- 2023
- Full Text
- View/download PDF
9. Sirtuin 2 inhibition modulates chromatin landscapes genome-wide to induce senescence in ATRX-deficient malignant glioma
- Author
-
Prit Benny Malgulwar, Carla Danussi, Sharvari Dharmaiah, William E. Johnson, Arvind Rao, and Jason T. Huse
- Subjects
Article - Abstract
Inactivating mutations inATRXcharacterize large subgroups of malignant gliomas in adults and children. ATRX deficiency in glioma induces widespread chromatin remodeling, driving transcriptional shifts and oncogenic phenotypes. Effective strategies to therapeutically target these broad epigenomic sequelae remain undeveloped. We utilized integrated mulit-omics and the Broad Institute Connectivity Map (CMAP) to identify drug candidates that could potentially revert ATRX-deficient transcriptional changes. We then employed disease-relevant experimental models to evaluate functional phenotypes, coupling these studies with epigenomic profiling to elucidate molecular mechanim(s). CMAP analysis and transcriptional/epigenomic profiling implicated the Class III HDAC Sirtuin2 (Sirt2) as a central mediator of ATRX-deficient cellular phenotypes and a driver of unfavorable prognosis in ATRX-deficient glioma. Sirt2 inhibitors reverted Atrx-deficient transcriptional signatures in murine neuroprogenitor cells (mNPCs) and impaired cell migration in Atrx/ATRX-deficient mNPCs and human glioma stem cells (GSCs). While effects on cellular proliferation in these contexts were more modest, markers of senescence significantly increased, suggesting that Sirt2 inhibition promotes terminal differentiation in ATRX-deficient glioma. These phenotypic effects were accompanied by genome-wide shifts in enhancer-associated H3K27ac and H4K16ac marks, with the latter in particular demonstrating compelling transcriptional links to Sirt2-dependent phenotypic reversals. Motif analysis of these data identified the transcription factor KLF16 as a mediator of phenotype reversal in Atrx-deficient cells upon Sirt2 inhibition. Finally, Sirt2 inhibition impaired growth and increased senescence in ATRX-deficient GSCsin vivo. Our findings indicate that Sirt2 inhibition selectively targets ATRX-deficient gliomas through global chromatin remodeling, while demonstrating more broadly a viable approach to combat complex epigenetic rewiring in cancer.Graphical AbstractOne Sentence SummaryOur study demonstrates that SIRT2 inhibition promotes senescence in ATRX-deficient glioma model systems through global epigenomic remodeling, impacting key downstream transcriptional profiles.
- Published
- 2023
- Full Text
- View/download PDF
10. EPCO-23. DYSFUNCTION OF LARGE H3K9ME3 DOMAINS IN ATRX DEFICIENT GLIOMAS INDUCES GENETIC REARRANGEMENTS AND LATENT DEVELOPMENTAL SIGNALING NETWORKS THROUGH SUPER-ENHANCER LANDSCAPES
- Author
-
Prit Benny Malgulwar, Carla Danussi, Anand Singh, Ajay Kumar Saw, Kunal Rai, and Jason Huse
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
Somatic alterations in ATRX (Alpha Thalassemia/Mental Retardation Syndrome X-linked), a member of SWI/SNF family chromatin regulator has been found to be frequently mutated in diffuse gliomas and defines molecular subtypes with aggressive behavior. Mechanistically, ATRX regulates incorporation of histone H3.3 into chromatin sites across the genome, maintains alternative lengthening of telomeres and establishes genomic distribution of polycomb responsive genes. To understand in depth role of ATRX in gliomas, we performed ChIP-seq and/or Cut-and-tag for histone marks that define active transcription, enhancers, repressors, gene bodies and CTCF on Atrx intact and deficient mNPCs (mouse Neural Progenitor Cells). Our integrated analysis reports depletion of H3K9me3 loci’s that coincidently enriched with Atrx binding sites and Lamina-Associated Domains with genes enriched for cell cycle, motility and chromosome organization. This chromatin perturbations at heterochromatin domains was further confirmed in our Hi-C analysis with switching of A-B and B-A compartments, reorganization of TADs with occasional CTCF marks and gain of novel interacting loops that showed gene expression leakage required for gliomagenesis using Capture Hi-C. Notably, we observed aberrant levels of endogenous retroviral elements (ERVs) family members, including upregulation of Line-1 elements in mNPCs and patient derived glioma stem cells (GSCs) and our analysis shows increased copy number variations in ATRX deficient gliomas as a consequence of Line-1 activation in these subsets of tumors. Finally, our integrated omics- analysis demonstrates enrichment of super-enhancers in Atrx deficient background with several putative druggable candidates for clinical benefits. As an example, we show presence of H3K27ac super-enhancer over HOXA locus and targeting pan-HOXA genes using small inhibitor peptides diminished proliferation and migration of mNPCs and GSCs with increased in cell apoptosis with alterations in downstream developmental signaling pathways. To summarize,our data establishes tangible links between Atrx deficiency and multiscale dysregulated cellular phenotype in gliomas with identifying novel targets for therapy.
- Published
- 2022
- Full Text
- View/download PDF
11. ATRX loss in glioma results in dysregulation of cell-cycle phase transition and ATM inhibitor radio-sensitization
- Author
-
Tingting Qin, Brendan Mullan, Ramya Ravindran, Dana Messinger, Ruby Siada, Jessica R. Cummings, Micah Harris, Ashwath Muruganand, Kalyani Pyaram, Zachary Miklja, Mary Reiber, Taylor Garcia, Dustin Tran, Carla Danussi, Jacqueline Brosnan-Cashman, Drew Pratt, Xinyi Zhao, Alnawaz Rehemtulla, Maureen A. Sartor, Sriram Venneti, Alan K. Meeker, Jason T. Huse, Meredith A. Morgan, Pedro R. Lowenstein, Maria G. Castro, Viveka Nand Yadav, and Carl Koschmann
- Subjects
Male ,X-linked Nuclear Protein ,Brain Neoplasms ,Cell Cycle ,Primary Cell Culture ,Cell Cycle Checkpoints ,Glioma ,General Biochemistry, Genetics and Molecular Biology ,Isocitrate Dehydrogenase ,Article ,Histones ,Mice, Inbred C57BL ,Mice ,Cell Line, Tumor ,Checkpoint Kinase 1 ,Mutation ,Animals ,Humans ,Female ,Neoplasm Recurrence, Local - Abstract
ATRX, a chromatin remodeler protein, is recurrently mutated in H3F3A-mutant pediatric glioblastoma (GBM) and isocitrate dehydrogenase (IDH)-mutant grade 2/3 adult glioma. Previous work has shown that ATRX-deficient GBM cells show enhanced sensitivity to irradiation, but the etiology remains unclear. We find that ATRX binds the regulatory elements of cell-cycle phase transition genes in GBM cells, and there is a marked reduction in Checkpoint Kinase 1 (CHEK1) expression with ATRX loss, leading to the early release of G2/M entry after irradiation. ATRX-deficient cells exhibit enhanced activation of master cell-cycle regulator ATM with irradiation. Addition of the ATM inhibitor AZD0156 doubles median survival in mice intracranially implanted with ATRX-deficient GBM cells, which is not seen in ATRX-wild-type controls. This study demonstrates that ATRX-deficient high-grade gliomas (HGGs) display Chk1-mediated dysregulation of cell-cycle phase transitions, which opens a window for therapies targeting this phenotype.
- Published
- 2021
12. EPCO-08. ATRX DEFICIENCY INDUCES DYSFUNCTIONAL HETEROCHROMATIN ARCHITECTURE IN GLIOMAS AND ESTABLISHES DISEASE-DEFINING TRANSCRIPTIONAL NETWORKS
- Author
-
Anand K Singh, Kasthuri Kannan, Carla Danussi, Prit Benny Malgulwar, Kunal Rai, and Jason T. Huse
- Subjects
Cancer Research ,biology ,Heterochromatin ,Epigenome ,26th Annual Meeting & Education Day of the Society for Neuro-Oncology ,medicine.disease ,Phenotype ,Chromatin ,Telomere ,Cell biology ,Alpha-thalassemia mental retardation syndrome ,Histone ,Oncology ,medicine ,biology.protein ,Neurology (clinical) ,ATRX - Abstract
Loss of ATRX (Alpha Thalassemia/Mental Retardation Syndrome X, a member of SWI/SNF family chromatin regulator is altered in diffuse gliomas and defines molecular subtypes with aggressive behavior. Mechanistically, ATRX regulates incorporation of histone H3.3 into chromatin sites across the genome, maintains alternative lengthening of telomeres and establishes genomic distribution of polycomb responsive genes. We have recently reported Atrx deficiency induces glioma oncogenic features via widespread alterations in chromatin accessibility using mouse Neural Progenitor Cells (mNPCs- Tp53 -/-,Atrx -/-). Surprisingly, Atrx was found to be associated with transcription start site and enhancer regions, suggesting their strong association with epigenome architecture. In this background, we have recently performed ChIP-seq for histone marks that define active transcription, enhancers, repressors and gene bodies and Cohesion molecules on Atrx intact and deficient mNPCs. Our integrated analysis reports depletion of H3K9me3 loci’s with enrichment of H3K27me3 marks that coincidently enriched with Atrx binding sites and Lamina-Associated Domains (LADs). GSEA confirmed that the genes corresponding to “newly formed LADs” in mNPC-to-astrocyte differentiation are significantly enriched for genes down-regulated in Atrx deficient mNPCs and belongs to Gene Ontology categories such as cell cycle, chromosome organization and DNA damage. Alternatively, genes corresponding to decreased LAD association are enriched for up-regulated genes in Atrx deficient mNPCs and for regulation of differentiation, adhesion and cell death. Additionally, whole-genome bisulphite sequencing further demonstrated loss of methylation marks at H3K9me3 sites in Atrx deficient mNPCs, suggesting perturbations of heterochromatin regions leading to activation of canonical signals that define glioma phenotype and disease-state. To summarize, our data establishes tangible links between Atrx deficiency and dysregulated chromatin and heterochromatin architecture in gliomas and suggests the role of Atrx in establishing global chromatin features and transcriptional networks. Further, our data unravel novel therapeutic molecules/pathways for engineering potential.
- Published
- 2021
- Full Text
- View/download PDF
13. ATRX Loss in Glioma Results in Epigenetic Dysregulation of Cell Cycle Phase Transition
- Author
-
Micah Harris, Brendan Mullan, Jacqueline A. Brosnan-Cashman, Carla Danussi, Taylor Garcia, Carl Koschmann, Alnawaz Rehemtulla, Mary Reiber, Zachary Miklja, Sriram Venneti, Kalyani Pyaram, Xinyi Zhao, Tingting Qin, Jessica R. Cummings, Drew Pratt, Dustin Tran, Maureen A. Sartor, Ashwath Muruganand, Ramya Ravindran, Meredith A. Morgan, Alan K. Meeker, Viveka Nand Yadav, Maria G. Castro, Pedro R. Lowenstein, Ruby Siada, and Jason T. Huse
- Subjects
Chemistry ,Glioma ,Cancer research ,medicine ,Cell cycle phase transition ,Epigenetics ,CHEK1 ,Cell cycle ,medicine.disease ,ATRX ,Chromatin ,Cell cycle phase - Abstract
ATRX, a chromatin remodeler protein, is recurrently mutated in H3F3A-mutant pediatric glioblastoma (GBM) and IDH-mutant grade 2/3 adult glioma. Previous work has shown that ATRX-deficient GBM cells show enhanced sensitivity to irradiation, but the etiology remains unclear. We found that ATRX binds regulatory elements of cell cycle phase transition genes in GBM cells, and there is a marked reduction in Checkpoint Kinase 1 (CHEK1) expression with ATRX loss, leading to early release of G2/M entry after irradiation. ATRX-deficient cells exhibit enhanced activation of master cell cycle regulator ATM with irradiation. Addition of the ATM inhibitor AZD0156 doubles median survival in mice intra-cranially implanted with ATRX-deficient GBM cells, which is not seen in ATRX-wildtype controls. This study demonstrates that ATRX-deficient high-grade gliomas (HGGs) display epigenetic dysregulation of cell cycle phase transitions, which opens a new window for therapies targeting this unique phenotype.
- Published
- 2021
- Full Text
- View/download PDF
14. Identification of patient-derived glioblastoma stem cell (GSC) lines with the alternative lengthening of telomeres phenotype
- Author
-
Vladislav G. Sharin, Ravesanker Ezhilarasan, Carla Danussi, Christian Alvarez, Jie Yang, Erik P. Sulman, Ahsan Farooqi, David M. Irvin, Sharvari Dharmaiah, and Jason T. Huse
- Subjects
X-linked Nuclear Protein ,Biology ,lcsh:RC346-429 ,Pathology and Forensic Medicine ,Cellular and Molecular Neuroscience ,Mice ,Text mining ,Cell Line, Tumor ,medicine ,Animals ,Humans ,RNA, Messenger ,Telomerase ,Letter to the Editor ,lcsh:Neurology. Diseases of the nervous system ,business.industry ,Brain Neoplasms ,RNA ,Telomere ,medicine.disease ,Phenotype ,Cell culture ,Cancer research ,Neoplastic Stem Cells ,Identification (biology) ,Neurology (clinical) ,Stem cell ,business ,Glioblastoma - Published
- 2019
15. Local inhibition of elastase reduces EMILIN1 cleavage reactivating lymphatic vessel function in a mouse lymphoedema model
- Author
-
Eliana Pivetta, Bruna Wassermann, Paola Spessotto, Giulia Bosisio, Teresa Maria Elisa Modica, Alfonso Colombatti, Vincenzo Canzonieri, Orlando Maiorani, Carla Danussi, Lisa Del Bel Belluz, Francesco Boccardo, Pivetta, Eliana, Wassermann, Bruna, Belluz Del Bel, Lisa, Danussi, Carla, Modica, Teresa Maria, Maiorani, Orlando, Bosisio, Giulia, Boccardo, Francesco, Canzonieri, Vincenzo, Colombatti, Alfonso, and Spessotto, Paola
- Subjects
0301 basic medicine ,extracellular matrix ,government.form_of_government ,Proteinase Inhibitory Proteins, Secretory ,S46 ,human lymphoedematous tissue ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Lymphatic vessel ,medicine ,Animals ,Humans ,Lymphedema ,EMILIN1 ,neutrophil elastase ,secondary lymphoedema ,sivelestat ,Cells, Cultured ,Lymphatic Vessels ,Original Paper ,S50 ,Membrane Glycoproteins ,biology ,business.industry ,Sivelestat ,Lymph Leakage ,Endothelial Cells ,General Medicine ,Original Papers ,Mice, Inbred C57BL ,Disease Models, Animal ,Lymphatic Endothelium ,030104 developmental biology ,Lymphatic system ,medicine.anatomical_structure ,Neutrophil Infiltration ,chemistry ,S39 ,Neutrophil elastase ,Immunology ,biology.protein ,government ,Cancer research ,Female ,Lymph ,business - Abstract
Lymphatic vasculature critically depends on the connections of lymphatic endothelial cells with the extracellular matrix (ECM), which are mediated by anchoring filaments (AFs). The ECM protein EMILIN1 is a component of AFs and is involved in the regulation of lymphatic vessel functions: accordingly, Emilin1−/− mice display lymphatic vascular morphological alterations, leading to functional defects such as mild lymphoedema, lymph leakage and compromised lymph drainage. In the present study, using a mouse post-surgical tail lymphoedema model, we show that the acute phase of acquired lymphoedema correlates with EMILIN1 degradation due to neutrophil elastase (NE) released by infiltrating neutrophils. As a consequence, the intercellular junctions of lymphatic endothelial cells are weakened and drainage to regional lymph nodes is severely affected. The local administration of sivelestat, a specific NE inhibitor, prevents EMILIN1 degradation and reduces lymphoedema, restoring a normal lymphatic functionality. The finding that, in human secondary lymphoedema samples, we also detected cleaved EMILIN1 with the typical bands of an NE-dependent pattern of fragmentation establishes a rationale for a powerful strategy that targets NE inhibition. In conclusion, the attempts to block EMILIN1 degradation locally represent the basis for a novel ‘ECM’ pharmacological approach to assessing new lymphoedema treatments.
- Published
- 2016
- Full Text
- View/download PDF
16. CBIO-03. ATRX LOSS IN GLIOMA RESULTS IN EPIGENETIC DYSREGULATION OF CELL CYCLE PHASE TRANSITION
- Author
-
Carl Koschmann, Brendan Mullan, Viveka Nand Yadav, Jackie Brosnan-Cashman, Kalyani Pyaram, Drew Pratt, Ruby Siada, Maureen A. Sartor, Alan K. Meeker, Ramya Ravindran, Dustin Tran, Ashwat Muruganand, Taylor Garcia, Tingting Qin, Xinyi Zhao, Jason T. Huse, Maria G. Castro, Pedro R. Lowenstein, Mary Reiber, Meredith A. Morgan, Sriram Venneti, Alnawaz Rehemtulla, Chase Thomas, Carla Danussi, Zachary Miklja, and Micah Harris
- Subjects
Cancer Research ,Mutation ,Chemistry ,Cell cycle ,medicine.disease ,medicine.disease_cause ,Phenotype ,Chromatin ,Oncology ,Glioma ,medicine ,Cell cycle phase transition ,Cancer research ,Neurology (clinical) ,Epigenetics ,Cell Biology (Cell Cycle Regulation, DNA Repair/Modulation) ,ATRX - Abstract
Gliomas are a leading cause of cancer mortality in children and adults, and new targeted therapies are desperately needed. ATRX is a chromatin remodeling protein that is recurrently mutated in H3F3A-mutant pediatric glioblastoma (GBM) and IDH-mutant grade 2/3 adult glioma. We previously showed that loss of ATRX in glioma results in tumor growth and additional tumor mutations. However, the mechanism driving these phenotypes has not been fully established. We found that in ChIP-Seq/ChIP-qPCR of mouse neuronal precursor cells (NPCs) and GBM cells with isogenic ATRX loss, ATRX binds regulatory elements for cell cycle phase transition gene sets, and ATRX loss subsequently results in reduced expression. Furthermore, human GBM cells with ATRX knock-out demonstrate higher rates of cells in S and G2/M phases, with clusters of cells demonstrating reduced expression of cell cycle regulatory gene sets by single-cell sequencing (scSeq) analysis. In human and mouse GBM in vitro models, ATRX-deficient cells exhibit a seven-fold increase in mitotic index at 16 hours after sub-lethal radiation and enhanced activation of the master cell cycle regulator ATM with radiation. Treatment of ATRX-deficient gliomas with ATM inhibitors results in a selective increase in dysfunctional cell cycling and increased radio-sensitization in ATRX-deficient glioma cells. Using an ATM-luciferase reporter in orthotopically-implanted human GBM cells, both AZD0156 and AZD1390 demonstrate in vivo pathway inhibition. Mice intra-cranially implanted with ATRX-deficient GBM cells demonstrate a doubling of median survival compared to radiated controls (p=0.0018) when treated with AZD0156 combined with radiation; this is not seen in ATRX-sufficient models. This study demonstrates that ATRX-deficient high-grade gliomas display epigenetic dysregulation of cell cycle phase transitions, which opens a new window for therapies targeting this unique phenotype.
- Published
- 2020
- Full Text
- View/download PDF
17. GENE-37. ATRX INACTIVATION DISRUPTS GLOBAL HETEROCHROMATIN LANDSCAPES AND ALTERS DISEASE-RELEVANT TRANSCRIPTIONAL ACTIVITY
- Author
-
Kunal Rai, Grant M. Fischer, David J. Picketts, Anand K Singh, Jason T. Huse, Carla Danussi, and Kasthuri Kannan
- Subjects
Cancer Research ,Transcriptional activity ,Abstracts ,Oncology ,Heterochromatin ,Neurology (clinical) ,Disease ,Biology ,Gene ,ATRX ,Cell biology - Abstract
Diffusely infiltrating gliomas feature loss-of function mutations in the SWI/SNF chromatin remodeler gene ATRX as defining molecular alterations delineating major adult and pediatric disease subtypes. So far, ATRX inactivation in cancer has been mainly correlated with alternative lengthening of telomeres, however, we recently reported that Atrx deficiency drives glioma-relevant phenotypes, such as increased motility and astrocytic differentiation profiles, by directly modulating epigenomic landscapes and the corresponding transcriptional profiles in glioma cells of origin. In particular, Atrx deficiency was associated with disruptions in H3.3 histone content at key genetic loci. To further understand the downstream epigenomic dysfunction induced by ATRX deficiency, we compared genome-wide chromatin-state maps of Atrx+ and Atrx- primary murine neuroepithelial progenitors (mNPCs). This ChIP–seq analysis revealed major differences in the localization of heterochromatin repressive marks H3K9me3 and H3K27me3. Specifically, we identified peculiar locations in the genome displaying H3K9me3 depletion and gain of H3K27me3 upon Atrx inactivation. Interestingly, these regions were flanked by Atrx binding sites and perfectly co-localized with Lamina-Associated Domains (LADs). LADs are widely involved in the control of gene expression programs during lineage commitment and terminal differentiation; typically they have a cell-specific distribution and are very dynamic during differentiation stages. Gene Set Enrichment Analysis confirmed that the genes corresponding to newly formed LADs in mNPC-to-astrocyte differentiation are significantly enriched for genes down-regulated in Atrx deficient mNPCs and belonging to Gene Ontology categories such as cell cycle, chromosome organization and DNA damage. On the other hand, genes corresponding to decreased LAD association are enriched for up-regulated genes in Atrx- mNPCs and for regulation of differentiation, adhesion and cell death. These data are in perfect agreement with our previously described glioma-relevant phenotypes associated with Atrx deficiency and indicate a novel role of Atrx in regulating the spatial organization of heterochromatin and its underlying transcriptional activity.
- Published
- 2018
18. G-quadruplex DNA drives genomic instability and represents a targetable molecular abnormality in ATRX-deficient malignant glioma
- Author
-
Rachna Shah, Timothy A. Chan, Jason T. Huse, Gregory J. Riggins, Carla Danussi, Jie Yang, Kasthuri Kannan, Wei H. Wu, Erik P. Sulman, and Yuxiang Wang
- Subjects
Genome instability ,0303 health sciences ,Programmed cell death ,DNA damage ,Context (language use) ,Biology ,Molecular Abnormality ,medicine.disease ,3. Good health ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,chemistry ,030220 oncology & carcinogenesis ,Glioma ,Cancer research ,medicine ,DNA ,ATRX ,030304 developmental biology - Abstract
Mutational inactivation ofATRX(α-thalassemia mental retardation X-linked) represents a defining molecular alteration in large subsets of malignant glioma. Yet the pathogenic consequences of ATRX deficiency remain unclear, as do tractable mechanisms for its therapeutic targeting. Here we report that ATRX loss in isogenic glioma model systems induces replication stress and DNA damage by way of G-quadruplex (G4) DNA secondary structure. Moreover, these effects are associated with the acquisition of disease-relevant copy number alterations over time. We then demonstrate, bothin vitroandin vivo, that ATRX deficiency selectively enhances DNA damage and cell death following chemical G4 stabilization. Finally, we show that G4 stabilization synergizes with other DNA-damaging therapies, including ionizing radiation, in the ATRX-deficient context. Our findings reveal novel pathogenic mechanisms driven by ATRX deficiency in glioma, while also pointing to tangible strategies for drug development.
- Published
- 2018
- Full Text
- View/download PDF
19. GENE-24. ATRX DEFICIENCY IN GLIOMA CELLS OF ORIGIN PROMOTES DISEASE-DEFINING PHENOTYPES BY WAY OF GLOBAL EPIGENOMIC REMODELING
- Author
-
Chad J. Creighton, David J. Picketts, Kasthuri Kannan, Frederick Lang, Jason T. Huse, Van Arnam J, Promita Bose, Carla Danussi, E.P. Sulman, Tang O, Adriana Heguy, Miller C, Timothy A. Chan, Pedro Silberman, Benjamin Deneen, Mark Vitucci, and Prasanna Tamarapu Parthasarathy
- Subjects
Cancer Research ,Disease ,Computational biology ,Biology ,medicine.disease ,Phenotype ,Abstracts ,Oncology ,Glioma ,medicine ,Neurology (clinical) ,Gene ,ATRX ,Epigenomics - Abstract
Epigenomic abnormalities are increasingly recognized as driver mechanisms in cancer, and somatic mutations in genes encoding epigenetic regulators have now been widely reported across a number of cancer types. Diffusely infiltrating gliomas, feature loss-of function mutations in the SWI/SNF chromatin remodeler gene ATRX as defining molecular alterations delineating major adult and pediatric disease subtypes. Within this disease context, ATRX deficiency invariably co-occurs with mutations in TP53 and in genes encoding either IDH1/2 in adults or H3.3 histone in children. So far, ATRX deficiency has been linked to a wide spectrum of physiological dysfunction, including aberrant gene regulation, abnormal telomere maintenance, genomic instability, and aneuploidy, but the molecular mechanism(s) by which ATRX deficiency promotes oncogenesis are still largely unknown, particularly those involving epigenomic dysregulation. To model these events in putative glioma cells of origin, we inactivated Atrx in primary murine neuroepithelial progenitors (mNPCs). Interestingly, Atrx loss induced NPCs to stop growing as neurospheres and adopt an adherent phenotype, while also exhibiting significantly increased motility. Moreover, when cultured in differentiation conditions, Atrx deficient cells displayed upregulation of astrocyte markers and downregulation of neuronal and oligodendrocyte markers, suggesting that Atrx directly regulates NPCs differentiation state and potential. Notably, the observed phenotypes correlated with altered gene expression profiles involving molecular networks implicated in development, regulation of signal transduction and cellular motility and invasion. Integrating these transcriptional changes with shifts in chromatin accessibility occurring with Atrx deficiency and genome-wide Atrx distribution, as determined by ChIP-seq, revealed highly significant spatial correlations between differentially expressed genes, regions of altered chromatin compaction, histone composition and genomic sites normally occupied by Atrx. Taken together, these findings demonstrate that Atrx deficiency induces widespread disruptions in chromatin organization, which in turn lead to dramatic shifts in gene expression and acquisition of disease-relevant phenotypes in putative glioma cells of origin.
- Published
- 2017
20. GENE-17. ATRX LOSS IN GLIOMA RESULTS IN EPIGENETIC DYSREGULATION OF THE G2/M CHECKPOINT AND SENSITIVITY TO ATM INHIBITION
- Author
-
Ruby Siada, Ashwath Muruganand, Ramya Ravindran, Tingting Qin, Viveka Nand Yadav, Carl Koschmann, Pedro R. Lowenstein, Alnawaz Rehemtulla, Carla Danussi, Xinyi Zhao, Alan K. Meeker, Maureen A. Sartor, Meredith A. Morgan, Sriram Venneti, Maria G. Castro, Drew Pratt, Jason T. Huse, Micah Harris, Jacqueline A. Brosnan-Cashman, Taylor Garcia, and Brendan Mullan
- Subjects
Genetics and Epigenetics ,Cancer Research ,Mutation ,Cell cycle checkpoint ,Biology ,Cell cycle ,medicine.disease ,medicine.disease_cause ,Chromatin ,Oncology ,Glioma ,medicine ,Cancer research ,Neurology (clinical) ,Epigenetics ,Gene ,ATRX - Abstract
Gliomas are a leading cause of cancer mortality in children and adults and new targeted therapies are desperately needed. ATRX is a chromatin remodeling protein that is recurrently mutated in H3F3A-mutant pediatric GBM and IDH-mutant grade 2/3 adult glioma. We previously showed that loss of ATRX in glioma results in tumor growth and additional tumor mutations. However, the mechanism driving these phenotypes has not been fully established. We found that in ChIP-Seq datasets of mouse neuronal precursor cells (NPCs) and experimental models of human glioma cells, ATRX binds and regulates the chromatin state of promoters and enhancers for gene sets associated with regulation of the cell cycle G2/M checkpoint. In line with this, analysis of single-cell seq (sc-seq) data from IDH-mutant gliomas (n=16) shows that ATRX-mutant tumors (IDH-A) demonstrate a population of cycling cells with dysregulated cell cycle phase gene set expression when compared to ATRX-wildtype tumors (IDH-O). In glioma models, ATRX-deficient cells exhibit a seven-fold increase in mitotic index at 16 hours after sub-lethal radiation and enhanced activation of the master cell cycle regulator ATM with radiation. Treatment of ATRX-deficient gliomas with ATM inhibitors results in a selective increase in dysfunctional cell cycling and increased radio-sensitization in ATRX-deficient glioma cells. Using an ATM-luciferase reporter in orthotopically-implanted human GBM cells, both AZD0156 and AZD1390 demonstrate in vivo pathway inhibition. Mice intra-cranially implanted with ATRX-deficient GBM cells demonstrate a doubling of median survival compared to radiated controls (p=0.0018) when treated with AZD0156 combined with radiation. This study demonstrates that ATRX-deficient glioma display epigenetic dysregulation of the G2/M checkpoint, which opens a new window for therapies targeting this unique phenotype.
- Published
- 2019
- Full Text
- View/download PDF
21. ATRX Mutant Glioblastoma Stem Cell (GSC) Lines with the Alternative Lengthening of Telomeres Phenotype Display Sensitivity to G-Quadruplex Stabilization
- Author
-
Christian Alvarez, J. Yang, S. Dharmiah, Erik P. Sulman, D. Irwin, Jason T. Huse, Ahsan Farooqi, Carla Danussi, Ravesanker Ezhilarasan, and Vladislav G. Sharin
- Subjects
Cancer Research ,Radiation ,business.industry ,Mutant ,G-quadruplex ,medicine.disease ,Phenotype ,Cell biology ,Telomere ,Oncology ,Medicine ,Radiology, Nuclear Medicine and imaging ,Stem cell ,business ,ATRX ,Glioblastoma - Published
- 2019
- Full Text
- View/download PDF
22. HGG-08. ATRX LOSS IN PEDIATRIC GBM RESULTS IN EPIGENETIC DYSREGULATION OF G2/M CHECKPOINT MAINTENANCE AND SENSITIVITY TO ATM INHIBITION
- Author
-
Maria G. Castro, Ruby Siada, Taylor Garcia, Meredith A. Morgan, Drew Pratt, Tingting Qin, Jason T. Huse, Xinyi Zhao, Alan K. Meeker, Alnawaz Rehemtulla, Jacqueline A. Brosnan-Cashman, Pedro R. Lowenstein, Carl Koschmann, Brendan Mullan, Sriram Venneti, Carla Danussi, and Viveka Nand Yadav
- Subjects
Cancer Research ,Cell cycle checkpoint ,biology ,business.industry ,Cell cycle ,medicine.disease ,Histone ,Oncology ,CpG site ,Tumor progression ,Glioma ,biology.protein ,medicine ,Cancer research ,Neurology (clinical) ,Epigenetics ,High Grade Glioma ,business ,ATRX - Abstract
ATRX is a histone chaperone protein recurrently mutated in pediatric GBM. We previously confirmed its role in tumor progression and mutational burden in glioma. However, the mechanism which mediates the proliferative advantage of ATRX loss in pediatric GBM remains unexplained. Recent data revealed a distinct pattern of DNA binding sites of the ATRX protein using ChIP-seq in mouse neuronal precursor cells (mNPCs). Using the ATRX peaks identified in p53(-/-) mNPCs, we confirmed that ATRX binding sites were significantly enriched in gene promoters (p
- Published
- 2019
- Full Text
- View/download PDF
23. EMILIN1/α9β1 Integrin Interaction Is Crucial in Lymphatic Valve Formation and Maintenance
- Author
-
Eliana Pivetta, Alfonso Colombatti, Roberto Doliana, Teresa Maria Elisa Modica, Bruna Wassermann, Lisa Del Bel Belluz, Paola Spessotto, Patrizia Sabatelli, Andrés F. Muro, and Carla Danussi
- Subjects
Integrins ,government.form_of_government ,Integrin ,Extracellular matrix ,Mice ,Cell Movement ,Interstitial fluid ,Animals ,Protein Interaction Maps ,Lymphangiogenesis ,Molecular Biology ,Cells, Cultured ,Cell Proliferation ,Lymphatic Vessels ,Mice, Knockout ,Membrane Glycoproteins ,biology ,Endothelial Cells ,EMILIN1 ,Articles ,Cell Biology ,Cell biology ,Mice, Inbred C57BL ,Endothelial stem cell ,Lymphatic Endothelium ,Lymphatic system ,Immunology ,biology.protein ,government - Abstract
Lymphatic vasculature plays a crucial role in the maintenance of tissue interstitial fluid balance. The role of functional collecting lymphatic vessels in lymph transport has been recently highlighted in pathologies leading to lymphedema, for which treatments are currently unavailable. Intraluminal valves are of paramount importance in this process. However, valve formation and maturation have not been entirely elucidated yet, in particular, the role played by the extracellular matrix (ECM). We hypothesized that EMILIN1, an ECM multidomain glycoprotein, regulates lymphatic valve formation and maintenance. Using a mouse knockout model, we show that in the absence of EMILIN1, mice exhibit defects in lymphatic valve structure and in lymph flow. By applying morphometric in vitro and in vivo functional assays, we conclude that this impaired phenotype depends on the lack of α9β1 integrin engagement, the specific lymphatic endothelial cell receptor for EMILIN1, and the ensuing derangement of cell proliferation and migration. Our data demonstrate a fundamental role for EMILIN1-integrin α9 interaction in lymphatic vasculature, especially in lymphatic valve formation and maintenance, and underline the importance of this ECM component in displaying a regulatory function in proliferation and acting as a "guiding" molecule in migration of lymphatic endothelial cells.
- Published
- 2013
- Full Text
- View/download PDF
24. The integrated landscape of driver genomic alterations in glioblastoma
- Author
-
Pietro Zoppoli, Alan X. Ji, Darell D. Bigner, Jeffrey N. Bruce, Francesco Niola, Gilbert G. Privé, Gaurav Gupta, Carla Danussi, Francesco Abate, Anna Lasorella, Vladimir Trifonov, Kenneth Aldape, David J. Pisapia, Angelica Castano, Stephen T. Keir, Marie Lia, Igor Dolgalev, Adriana Heguy, Peter Canoll, Antonio Iavarone, Roger E. McLendon, Paola Porrati, Tom Mikkelsen, Joseph M. Chan, Serena Pellegatta, Raul Rabadan, Veronique Frattini, Gaetano Finocchiaro, Hai Yan, Frattini, V., Trifonov, V., Chan, J. M., Castano, A., Lia, M., Abate, F., Keir, S. T., Ji, A. X., Zoppoli, P., Niola, F., Danussi, C., Dolgalev, I., Porrati, P., Pellegatta, S., Heguy, A., Gupta, G., Pisapia, D. J., Canoll, P., Bruce, J. N., Mclendon, R. E., Yan, H., Aldape, K., Finocchiaro, G., Mikkelsen, T., Prive, G. G., Bigner, D. D., Lasorella, A., Rabadan, R., and Iavarone, A.
- Subjects
Delta Catenin ,Somatic cell ,Biology ,medicine.disease_cause ,Article ,Loss of heterozygosity ,03 medical and health sciences ,0302 clinical medicine ,Glioma ,Genetics ,medicine ,Humans ,Copy-number variation ,Gene ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,Mutation ,Brain Neoplasms ,Catenins ,Genomics ,medicine.disease ,3. Good health ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,Cancer research ,Stem cell ,Glioblastoma ,Transcription Factors - Abstract
Glioblastoma is one of the most challenging forms of cancer to treat. Here we describe a computational platform that integrates the analysis of copy number variations and somatic mutations and unravels the landscape of in-frame gene fusions in glioblastoma. We found mutations with loss of heterozygosity in LZTR1, encoding an adaptor of CUL3-containing E3 ligase complexes. Mutations and deletions disrupt LZTR1 function, which restrains the self renewal and growth of glioma spheres that retain stem cell features. Loss-of-function mutations in CTNND2 target a neural-specific gene and are associated with the transformation of glioma cells along the very aggressive mesenchymal phenotype. We also report recurrent translocations that fuse the coding sequence of EGFR to several partners, with EGFR-SEPT14 being the most frequent functional gene fusion in human glioblastoma. EGFR-SEPT14 fusions activate STAT3 signaling and confer mitogen independence and sensitivity to EGFR inhibition. These results provide insights into the pathogenesis of glioblastoma and highlight new targets for therapeutic intervention. © 2013 Nature America, Inc. All rights reserved.
- Published
- 2013
- Full Text
- View/download PDF
25. CADD-27. G-QUADRUPLEX DNA DRIVES GENOMIC INSTABILITY AND REPRESENTS A TARGETABLE MOLECULAR ABNORMALITY IN ATRX-DEFICIENT MALIGNANT GLIOMA
- Author
-
Gregory J. Riggins, Kasthuri Kannan, Jie Yang, Yuxiang Wang, Rachna Shah, Jason T. Huse, Carla Danussi, Wei Wu, Timothy A. Chan, and Erik P. Sulman
- Subjects
Genome instability ,Cancer Research ,DNA damage ,Biology ,medicine.disease ,Molecular Abnormality ,Chromatin ,Abstracts ,chemistry.chemical_compound ,Oncology ,chemistry ,Glioma ,medicine ,Cancer research ,Neurology (clinical) ,Epigenetics ,ATRX ,DNA - Abstract
Mutational inactivation of ATRX (a-thalassemia mental retardation X-linked) represents a defining molecular alteration in large subsets of malignant glioma. ATRX encodes a chromatin binding protein widely implicated in epigenetic regulation and remodeling. However, multiple studies have also associated its loss in cancer with replication stress, DNA damage, and copy number alterations (CNAs). The mechanisms by which ATRX deficiency drives this global genomic instability remain unclear. Here we report that ATRX inactivation in isogenic glioma model systems induces replication stress and DNA damage by promoting the formation of deleterious G-quadruplex (G4) secondary structure in DNA. Moreover, these effects are associated with the acquisition of disease-relevant CNAs over time. Prior work has linked G4s with genomic instability as well as CNAs in cancer. We then demonstrate, both in vitro and in vivo, that chemical G4 stabilization with CX-3543 (Quarfloxin) selectively enhances cell death in ATRX deficient isogenic cell lines as well as ATRX-mutant primary glioma stem cells derived from patients. Finally, we show that G4 stabilization synergizes with other DNA-damaging therapies, including ionizing radiation, in the ATRX-deficient context. Our findings clarify distinct mechanisms by which DNA secondary structure influences ATRX-deficient glioma pathogenesis and indicate that G4-stabilization may represent and attractive therapeutic strategy for the selective targeting of ATRX-mutant cancers. Opportunities for the development of radiosensitization approaches based on G4-stabilization are particularly intriguing, as ionizing radiation remains among the most effective non-surgical treatments for malignant glioma.
- Published
- 2018
- Full Text
- View/download PDF
26. Abstract 4322: Atrx inactivation drives motility and dysregulates differentiation in glioma cells of origin through global epigenomic remodeling
- Author
-
C. Ryan Miller, Jason T. Huse, Oliver Y. Tang, Kasthuri Kannan, John S. Van Arnam, Carla Danussi, Timothy A. Chan, Promita Bose, Pedro Silberman, Adriana Heguy, Mark Vitucci, Benjamin Deneen, Frederick F. Lang, Chad J. Creighton, Erik P. Sulman, and David J. Picketts
- Subjects
Cancer Research ,Oncology ,Glioma ,medicine ,Motility ,Biology ,medicine.disease ,ATRX ,Epigenomics ,Cell biology - Abstract
Comprehensive genomic profiling in cancer continues to reveal frequent alterations in epigenetic regulators, firmly implicating chromatin biology in the oncogenic process. For instance, genetic inactivation of the SWI/SNF chromatin regulator ATRX (α-thalassemia mental retardation X-linked) represents a defining molecular alteration in both adult and pediatric malignant glioma, and occurs frequently in other cancers as well. ATRX deficiency has been linked to a wide spectrum of physiological dysfunction, including aberrant gene regulation, abnormal telomere maintenance, genomic instability, and aneuploidy. However, the precise oncogenic mechanism(s) induced by ATRX deficiency remain unclear, particularly those involving epigenomic dysregulation. To model these events in putative glioma cells of origin, we inactivated Atrx in primary murine neuroepithelial progenitors (mNPCs). Atrx loss, especially when coupled with Tp53 inactivation, promoted mNPC motility while also modulating differentiation state and potential, effectively recapitulating characteristic disease phenotypes and molecular features. Moreover, these phenotypes correlated with altered gene expression profiles in functionally relevant molecular networks (e.g. cell differentiation and migration). Integrating these transcriptional changes with shifts in chromatin accessibility occurring with Atrx deficiency, along with genome-wide Atrx distribution as determined by ChIP-seq, revealed highly significant spatial correlations between differentially expressed genes, regions of altered chromatin compaction, and genomic sites normally occupied by Atrx. Finally, target genes mediating specific Atrx-deficient phenotypes in vitro exhibited similarly selective misexpression in ATRX-mutant human glioma tissues and cell lines. These findings demonstrate that, in appropriate cellular and molecular contexts, ATRX deficiency and its epigenomic sequelae are sufficient to induce disease-defining oncogenic phenotypes. Citation Format: Carla Danussi, Promita Bose, Pedro Silberman, John S. Van Arnam, Mark Vitucci, Oliver Tang, Adriana Heguy, Timothy A. Chan, Erik P. Sulman, Frederick Lang, Chad J. Creighton, Benjamin Deneen, C Ryan Miller, David J. Picketts, Kasthuri Kannan, Jason T. Huse. Atrx inactivation drives motility and dysregulates differentiation in glioma cells of origin through global epigenomic remodeling [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 4322.
- Published
- 2018
- Full Text
- View/download PDF
27. Abstract 345: Characterizing and therapeutically targeting G-quadruplex DNA in ATRX-mutant glioma
- Author
-
Yuxiang Wang, Timothy A. Chan, Jason T. Huse, Kasthuri Kannan, and Carla Danussi
- Subjects
Cancer Research ,chemistry.chemical_compound ,Oncology ,chemistry ,Glioma ,Mutant ,medicine ,medicine.disease ,G-quadruplex ,Molecular biology ,ATRX ,DNA - Abstract
Inactivating ATRX mutations are defining molecular alterations in several cancer variants, including large subsets of malignant glioma. ATRX encodes a chromatin binding protein widely implicated in epigenetic and transcriptional regulation. However, ATRX is also thought to guard against genomic instability, in part by mitigating the formation of deleterious G-quadruplex (G4) DNA secondary structures. G4s have been shown to stall DNA replication leading to single-strand breaks and, potentially, DNA damage and genomic instability. To study this biology in a glioma-relevant context, we generated an isogenic astrocyte model featuring intact or deficient ATRX. While ATRX deficiency had no effect on baseline proliferation, cell cycle progression, and apoptosis, it led to markedly increased G4 formation, DNA damage, and replication stress signaling. These effects were reversed by ATRX re-expression. Moreover, ATRX-deficient astrocytes acquired copy number alterations over time, unlike ATRX-intact counterparts. Given these findings, we hypothesized that therapeutically targeting G4s might serve to exploit the inherent genomic vulnerabilities of ATRX-deficient cancer. Remarkably, treatment with a G4 stabilizing compound (CX3543) selectively inhibited growth and colony formation in ATRX-deficient astrocytes at nanomolar concentrations, and synergistically potentiated the effects of ionizing radiation and hydroxyurea. Finally, CX3543 significantly inhibited the growth of patient-derived, ATRX-deficient glioma xenografts in mice, with only minimal effects on ATRX-intact glioma xenografts. These results confirm and characterize a novel functionality for ATRX in the mitigation of genomic instability and point towards a viable strategy for therapeutically targeting ATRX deficiency in cancer. Citation Format: Yuxiang Wang, Carla Danussi, Kasthuri Kannan, Timothy A. Chan, Jason T. Huse. Characterizing and therapeutically targeting G-quadruplex DNA in ATRX-mutant glioma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 345.
- Published
- 2018
- Full Text
- View/download PDF
28. Emilin1 Deficiency Causes Structural and Functional Defects of Lymphatic Vasculature
- Author
-
Roberto Doliana, Patrizia Sabatelli, Alessandra Petrucco, Monica Montesi, Carla Danussi, Alfonso Colombatti, Giorgio M. Bressan, Bruna Wassermann, and Paola Spessotto
- Subjects
government.form_of_government ,Mice, Transgenic ,Biology ,Cell morphology ,Models, Biological ,lymphatic vessels ,Extracellular matrix ,Mice ,Elastic fibers ,Emilin1 ,lymphedema ,Animals ,Humans ,Lymphangiogenesis ,Lymph sacs ,Molecular Biology ,Cells, Cultured ,Mice, Inbred BALB C ,Membrane Glycoproteins ,Articles ,Cell Biology ,Extracellular Matrix ,Cell biology ,Mice, Inbred C57BL ,Lymphatic Endothelium ,Phenotype ,Lymphatic system ,Gene Expression Regulation ,Immunology ,Circulatory system ,government ,Lymph - Abstract
Lymphatic and blood vascular systems have distinct structural characteristics that reflect their specific and complementary functions. The lymphatic vasculature represents a second circulatory system and maintains tissue fluid homeostasis; it plays a major role in the absorption of dietary fat and in immune response, transporting lymphocytes and antigen-presenting cells to regional lymph nodes; finally, it provides routes for tumor metastasis (8). The lymphatic system consists of a complex network of lymphatic capillaries, which are uniquely adapted for the uptake of protein-rich lymph from tissue interstitium, and collecting lymphatic vessels that transport lymph back to the blood vascular system. The latter are surrounded by a basement membrane and smooth muscle cells, which are less organized than in blood vessels and, in addition, have intraluminal valves, which prevent lymph backflow (26). By contrast, lymphatic capillaries are blind-end vessels, lined by a single thin layer of overlapping lymphatic endothelial cells (LECs) directly connected to the surrounding extracellular matrix (ECM) by means of anchoring elastic filaments (18). These structures play a fundamental role in lymphatic-vessel function and represent one of the main distinguishing features between lymphatic and blood capillaries. When interstitial fluid pressure increases, anchoring filaments exert tension on LECs, thereby widening the capillary lumen and opening the overlapping cell junctions, which enable fluid and macromolecule uptake and cell entry. It is thought that abnormalities of anchoring filaments may reduce adsorption from the interstitium and propulsion of lymph and cells and promote pathological conditions, such as lymphedema or diseases related to impaired immune responses (9, 18). Furthermore, the perivascular ECM plays an integral role in lymphatic-vessel function, as the fluid equilibrium is controlled by the cooperation of both lymphatic function and the ECM (40). The elasticity and hydration of a tissue is determined by the composition and organization of the ECM. Extensive and chronic degradation of the ECM eventually renders lymphatic vessels nonresponsive to the changes in the interstitium and therefore causes dysfunction (28, 30). Lymphatic and blood endothelial cells express different lineage-specific molecules involved in the regulation of their biological functions that are frequently used as distinguishing markers, such as vascular endothelial growth factor receptor 3 (VEGFR-3) (22), podoplanin (3), Prox-1 (44), LYVE-1 (1, 34), neuropilin 2 (46), CCL21 (24), and desmoplakin (12). Recently, a comparative microarray analysis of gene expression profiles of lymphatic and blood endothelial cells identified previously unknown lymphatic lineage genes, including macrophage mannose receptor 1, plakoglobin, the chemokine CCL20, the integrin α9β1 (19, 32), and EMILIN1 (33). EMILIN1 is an ECM glycoprotein associated with elastic fibers (4, 6) and composed of an N-terminal cysteine-rich domain and the EMI domain (11), followed by a coiled-coil structure, a short collagenous stalk, and a C-terminal gC1q domain (7). EMILIN1 is particularly abundant in the walls of large blood vessels, such as the aorta (10), and has been implicated in multiple functions. EMILIN1 is involved in elastogenesis and in the maintenance of blood vascular cell morphology (48). It interacts with the α4β1 integrin through the gC1q1 domain (36) and has strong adhesive and migratory properties for different cell types (10, 36, 37). EMILIN1, via the EMI domain, regulates pro-transforming growth factor beta (TGF-β) maturation and is involved in blood pressure homeostasis (47). To directly investigate the physiological function of EMILIN1 in lymphatic vessels, we studied the effects of its absence in mice that had targeted deletions in the Emilin1 gene (48). Here, we report that EMILIN1 is highly expressed by LECs in vitro and that it colocalizes with lymphatic vessels in several mouse tissues. Importantly, Emilin1 deficiency results in hyperplasia and enlargement of lymphatic vessels and in a significant reduction of anchoring filaments compared to those of wild-type (WT) mice. The lymphatic vessels of Emilin1−/− mice are functionally altered. We found that lack of EMILIN1 leads to a mild lymphedema associated with inefficient lymph drainage and increased leakage. In addition, Emilin1−/− mice develop larger lymphangiomas than their WT littermates. Altogether, these findings demonstrate an important role of EMILIN1 in the structure-function relationship of lymphatic vessels and identify EMILIN1 as a lymphangiogenesis modulator.
- Published
- 2008
- Full Text
- View/download PDF
29. Laminin-332 (Laminin-5) is the major motility ligand for B cell chronic lymphocytic leukemia
- Author
-
Paola Spessotto, Bruna Wasserman, Carla Danussi, Antonella Zucchetto, Alfonso Colombatti, Vincenzo Canzonieri, Valter Gattei, Riccardo Bomben, Oriano Radillo, Roberto Perris, Massimo Degan, Spessotto, P, Zucchetto, A, Degan, M, Wasserman, B, Danussi, C, Bomben, R, Perris, R, Canzonieri, V, Radillo, O, Colombatti, A, and Gattei, V
- Subjects
Adult ,Male ,Pathology ,medicine.medical_specialty ,Immunoglobulin Variable Region ,Gene Expression ,Motility ,Extracellular matrix ,Cell Movement ,Laminin ,hemic and lymphatic diseases ,medicine ,Humans ,Protein Isoforms ,RNA, Messenger ,Cell adhesion ,Molecular Biology ,Lymph node ,Aged ,Aged, 80 and over ,biology ,Integrin alpha3beta1 ,Antibodies, Monoclonal ,Cell migration ,Middle Aged ,Immunohistochemistry ,Leukemia, Lymphocytic, Chronic, B-Cell ,Fibronectins ,Cell biology ,medicine.anatomical_structure ,Matrix Metalloproteinase 9 ,Mutation ,Reticular connective tissue ,biology.protein ,Matrix Metalloproteinase 2 ,Female ,Lymph Nodes ,Lymph ,Immunoglobulin Heavy Chains ,Cell Adhesion Molecules - Abstract
Cell adhesion and motility are central aspects in the pathophysiology of B cell chronic lymphocytic leukemia (B-CLL), but the role of specific extracellular matrix proteins is still to be completely unveiled. Purified peripheral blood neoplastic cells of B-CLL patients migrated poorly on laminins-111,-411,-511, but showed pronounced motility on laminin (LM)-332 in a high percentage of cases. B-CLL cell motility on LM-332 was mediated by the alpha 3 beta 1 integrin and was preferentially observed in cells carrying a mutated IgV(H) gene profile. Within normal lymph nodes, LM-332 was circumscribed around blood vessels and to areas corresponding to marginal zones, where it was deposited in a pattern reminiscent of reticular fibers. Conversely, in B-CLL involved lymph nodes, a positive LM-332 reticular mesh was diffusely evident, throughout the disrupted nodal architecture. In the present study we identified LM-332 as a crucial motility-promoting factor for B-CLL lymphocytes and as a potential constituent favoring the dissemination of B-CLL lymphocytes through vascular basement membranes and possibly lymph node compartments. (c) 2007 Elsevier B.V./International Society of Matrix Biology. All rights reserved.
- Published
- 2007
- Full Text
- View/download PDF
30. Neutrophil elastase-dependent cleavage compromises the tumor suppressor role of EMILIN1
- Author
-
Carla Danussi, Teresa Maria Elisa Modica, Paola Spessotto, Eliana Pivetta, Alfonso Colombatti, Vincenzo Canzonieri, Bruna Wassermann, and Lisa Del Bel Belluz
- Subjects
Integrins ,Neutrophils ,Proteolysis ,Integrin ,Extracellular matrix ,Cell Line, Tumor ,medicine ,Humans ,Genes, Tumor Suppressor ,Molecular Biology ,Tumor microenvironment ,Membrane Glycoproteins ,biology ,medicine.diagnostic_test ,Chemistry ,Elastase ,Proteolytic enzymes ,EMILIN1 ,Sarcoma ,Cell biology ,Extracellular Matrix ,Biochemistry ,Neutrophil elastase ,biology.protein ,Leukocyte Elastase ,Integrin alpha Chains - Abstract
Proteolysis of the extracellular matrix (ECM) is a key event in tumor growth and progression. The breakdown of ECM can lead to the generation of bioactive fragments that promote cell growth and spread. EMILIN1, a multidomain glycoprotein expressed in several tissues, exerts a crucial regulatory function through the engagement of α4/α9 integrins. Unlike the majority of ECM molecules that elicit a proliferative program, the signals emitting from EMILIN1 engaged by α4/α9β1 integrins are antiproliferative. In this study, aimed to demonstrate if the suppressor role of EMILIN1 was related to its structural integrity, we tested the possibility that EMILIN1 could be specifically cleaved. Among the proteolytic enzymes released in the tumor microenvironment we showed that neutrophil elastase cleaved EMILIN1 in three/four major fragments. The consequence of this proteolytic process was the impairment of its anti-proliferative role. Accordingly, EMILIN1 was digested in sarcomas and ovarian cancers. Sarcoma specimens were infiltrated by neutrophils (PMNs) and stained positively for elastase. The present findings highlight the peculiar activity of PMN elastase in disabling EMILIN1 suppressor function.
- Published
- 2013
31. RHPN2 drives mesenchymal transformation in malignant glioma by triggering RhoA activation
- Author
-
Antonio Iavarone, Anna Lasorella, Andreja Jovic, Uri David Akavia, Francesco Niola, Carla Danussi, and Dana Pe'er
- Subjects
Cancer Research ,RHOA ,DNA Copy Number Variations ,Cell Growth Processes ,medicine.disease_cause ,Article ,Mice ,Neural Stem Cells ,Cell Movement ,Glioma ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Adaptor Proteins, Signal Transducing ,Mutation ,biology ,Mesenchymal stem cell ,Cell migration ,Mesenchymal Stem Cells ,medicine.disease ,Molecular biology ,Phenotype ,Neural stem cell ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,HEK293 Cells ,Oncology ,Astrocytes ,Cancer research ,biology.protein ,Ectopic expression ,Glioblastoma ,rhoA GTP-Binding Protein ,Chromosomes, Human, Pair 19 - Abstract
Mesenchymal transformation is a hallmark of aggressive glioblastoma (GBM). Here, we report the development of an unbiased method for computational integration of copy number variation, expression, and mutation data from large datasets. Using this method, we identified rhophilin 2 (RHPN2) as a central genetic determinant of the mesenchymal phenotype of human GBM. Notably, amplification of the human RHPN2 gene on chromosome 19 correlates with a dramatic decrease in the survival of patients with glioma. Ectopic expression of RHPN2 in neural stem cells and astrocytes triggered the expression of mesenchymal genes and promoted an invasive phenotype without impacting cell proliferation. Mechanistically, these effects were implemented through RHPN2-mediated activation of RhoA, a master regulator of cell migration and invasion. Our results define RHPN2 amplification as a central genetic determinant of a highly aggressive phenotype that directs the worst clinical outcomes in patients with GBM. Cancer Res; 73(16); 5140–50. ©2013 AACR.
- Published
- 2013
32. GENT-41. CHARACTERIZING AND THERAPEUTICALLY TARGETING G-QUADRUPLEX DNA IN ATRX-MUTANT GLIOMA
- Author
-
Yuxiang Wang, Jason T. Huse, Adriana Heguy, Peter Lansdorp, and Carla Danussi
- Subjects
Cancer Research ,chemistry.chemical_compound ,Oncology ,chemistry ,Glioma ,Mutant ,medicine ,Neurology (clinical) ,medicine.disease ,G-quadruplex ,Molecular biology ,DNA ,ATRX - Published
- 2016
- Full Text
- View/download PDF
33. GENT-21. ATRX DEFICIENCY PROMOTES INVASION AND ALTERS THE DIFFERENTIATION CAPACITY OF GLIOMA CELLS OF ORIGIN BY DYSREGULATING GENOME-WIDE CHROMATIN ACCESSIBILITY AND MODULATING TRANSCRIPTIONAL PROGRAMS
- Author
-
Timothy A. Chan, Promita Bose, Sevin Turcan, Prasanna Tamarapu Parthasarathy, Pedro Silberman, Jason T. Huse, David J. Picketts, and Carla Danussi
- Subjects
Genetics ,Cancer Research ,Oncology ,Glioma ,medicine ,Neurology (clinical) ,Biology ,medicine.disease ,Genome ,ATRX ,Cell biology ,Chromatin - Published
- 2016
- Full Text
- View/download PDF
34. Cancer Stem Cells and the Microenvironment
- Author
-
Eliana Pivetta, Paola Spessotto, Carla Danussi, and Alfonso Colombatti
- Subjects
Tumor microenvironment ,Intravasation ,Biology ,medicine.disease_cause ,medicine.disease ,Primary tumor ,medicine.anatomical_structure ,Cancer stem cell ,Cancer cell ,Cancer research ,medicine ,Stem cell ,Carcinogenesis ,Lymph node - Abstract
The traditional interpretation of cancer progression is that tumor cells accumulate multiple genetic alterations, resulting in the capability to surmount a variety of obstacles including detachment from the primary tumor, intravasation into blood or lymphatic vessels, survival into circulation, extravasation and growth at a secondary site, lymph node and/or distant organ (Vogelstein and Kinzler, Nat Med 10:789–799, 2004). The hypothesis of a hierarchical organization of cells within at least certain tumors in which a subset of tumor cells has the property to self-renew, hence defined “stem-like” or cancer stem cells (CSC), and to generate and sustain the wide heterogeneity comprising the tumor is very attractive. However, the tumor cell-centered view of cancer progression ignores the contribution of the tumor microenvironment to the malignant phenotype. A tumor should be viewed as a complex dynamically evolving ecosystem where minor alterations may cause dramatic reorganization of the whole system and of the malignant versus benign behavior of cells. In fact, the tumor microenvironment is constantly changing and cancer cells, either CSC or more differentiated tumor cells, adapt, evolve, and survive during this process. Tumors grow in conducive microenvironments reminiscent of the normal stem cells niche concept. The tumor microenvironment is implicated in the transition from preinvasive to invasive growth and is regarded as a crucial participant in tumorigenesis promoting a more aggressive phenotype and has relevance also for therapeutical approaches.
- Published
- 2011
- Full Text
- View/download PDF
35. A newly generated functional antibody identifies Tn antigen as a novel determinant in cancer cell-lymphatic endothelium interaction
- Author
-
Fulvio Uggeri, Paola Spessotto, Alfonso Colombatti, Cristiana Campa, Carla Danussi, Anna Coslovi, Sergio Paoletti, Maria Teresa Mucignat, Danussi, C, Spessotto, P, MUCIGNAT M., T, Coslovi, A, Campa, C, Uggeri, F, Paoletti, Sergio, and Colombatti, A.
- Subjects
Monoclonal antibody ,Antibodies, Neoplasm ,medicine.drug_class ,government.form_of_government ,Tn antigen ,Mice, Nude ,Metastasi ,Biochemistry ,Malignant transformation ,Metastasis ,Mice ,Cell Line, Tumor ,Neoplasms ,Cell Adhesion ,medicine ,Animals ,Humans ,In vivo imaging ,Lymphatic endothelial cells ,Antigens, Tumor-Associated, Carbohydrate ,Endothelium ,Lymphatic endothelial cell ,Mice, Inbred BALB C ,Molecular Structure ,biology ,Antibodies, Monoclonal ,medicine.disease ,Immunohistochemistry ,Xenograft Model Antitumor Assays ,Molecular biology ,Lymphatic Endothelium ,Lymphatic system ,Lymphatic Metastasis ,Cancer cell ,biology.protein ,government ,Female ,Original Article ,Lymph Nodes ,Antibody - Abstract
Malignant transformation of epithelial cells is frequently associated with the alteration of glycosylation pathways. Tn is a common tumor-associated carbohydrate antigen present in 90% of human carcinomas and its expression correlates with metastatic potential and poor prognosis. Despite its relevance, the functional role of Tn in tumor biology has not been firmly established probably for the lack of appropriate experimental tools. Our aims were to produce highly reactive monoclonal antibodies against Tn making use of synthetically produced Tn and to test their usefulness for in vivo imaging as well as to define their potential functional activity in tumor cell spread. We immunized mice with Tn clustered on cationized BSA and screened the positive hybridomas with Tn-biotinylated alginate. Enzyme-linked immuno sorbent assay and immunofluorescence assays revealed that the most reactive anti-Tn IgM mAb (2154F12A4) selectively recognized Tn on the MCF7 breast cancer cell line since its binding to the cell membrane was completely abolished by preincubation with purified Tn. Importantly, QDot 800-conjugated mAb injected in MCF7-tumor bearing mice specifically bound to primary tumor lesions as well as to metastases in lymph nodes. In addition, this mAb was able to inhibit cancer cell adhesion to lymphatic endothelium suggesting a novel involvement of Tn in the lymphatic dissemination of cancer cells and hypothesizing future applications in inhibiting lymphatic metastases.
- Published
- 2009
36. EPIG-06ATRX DEFICIENCY PROMOTES MIGRATORY BEHAVIOR IN GLIOMA CELLS OF ORIGIN BY DYSREGULATING GENOME-WIDE CHROMATIN ACCESSIBILITY AND MODULATING TRANSCRIPTIONAL PROGRAMS
- Author
-
Timothy A. Chan, Prasanna Tamarapu Parthasarathy, Sevin Turcan, Carla Danussi, David J. Picketts, Jason T. Huse, and Promita Bose
- Subjects
Regulation of gene expression ,Genome instability ,Cancer Research ,Biology ,medicine.disease_cause ,Phenotype ,Chromatin ,Telomere ,Oncology ,medicine ,Cancer research ,Neurology (clinical) ,Epigenetics ,Carcinogenesis ,Abstracts from the 20th Annual Scientific Meeting of the Society for Neuro-Oncology ,ATRX - Abstract
Comprehensive genomic profiling in cancer continues to reveal frequent alterations in epigenetic regulators, firmly implicating chromatin biology in the oncogenic process. We and others recently found that inactivating mutations in the SWI/SNF family member ATRX represent defining molecular alterations in diffusely infiltrating gliomas, where they invariably pair with mutations in TP53 and IDH1/2. A number of studies have linked ATRX deficiency to a wide spectrum of physiological dysfunction, including aberrant gene regulation, abnormal telomere maintenance, genomic instability, and aneuploidy. Nevertheless, the molecular mechanism(s) by which ATRX deficiency promotes oncogenesis are still largely unestablished. To understand the role of Atrx loss-of-function mutations in gliomagenesis, we inactivated Atrx in Tp53-/- murine neuroepithelial progenitor cells (NPCs), which have been implicated as potential glioma cells of origin by several prior studies. Interestingly, Atrx loss induced NPCs to stop growing as neurospheres and adopt an adherent phenotype, while also exhibiting significantly increased cellular motility by transwell migration assay, the latter an established pathogenic feature of diffusely infiltrating gliomas. These phenotypes correlated with altered gene expression profiles involving molecular networks implicated in development, regulation of signal transduction, and cellular motility and invasion. Integrating these transcriptional changes with shifts in chromatin accessibility occurring with Atrx deficiency and genome-wide Atrx distribution, as determined by ChIP-seq, revealed highly significant spatial correlations between differentially expressed genes, regions of altered chromatin compaction, and genomic sites normally occupied by Atrx. Taken together, these findings demonstrate that Atrx deficiency induces widespread disruptions in chromatin organization, which in turn lead to dramatic shifts in gene expression and acquisition of disease-relevant phenotypes in putative glioma cells of origin. We are now in the process of identifying and characterizing the precise transcriptional and epigenetic events mediating the acquisition of Atrx-dependent behaviors in NPCs.
- Published
- 2015
- Full Text
- View/download PDF
37. EMILIN1 represents a major stromal element determining human trophoblast invasion of the uterine wall
- Author
-
Giada Monami, Oriano Radillo, Paola Spessotto, Roberta Bulla, Francesco Tedesco, Marta Cervi, Carla Danussi, Alfonso Colombatti, Fleur Bossi, Roberto Doliana, Spessotto, P, Bulla, Roberta, Danussi, C, Radillo, O, Cervi, M, Monami, G, Bossi, Fleur, Tedesco, Francesco, Doliana, R, and Colombatti, A.
- Subjects
medicine.medical_specialty ,Stromal cell ,Matrix Metalloproteinases, Membrane-Associated ,Placenta ,Integrin ,Connective tissue ,Integrin alpha4beta1 ,Matrix Metalloproteinase Inhibitors ,Cell Movement ,Pregnancy ,Internal medicine ,medicine ,Cell Adhesion ,Decidua ,Humans ,Neoplasm Invasiveness ,RNA, Small Interfering ,Cell adhesion ,Migration ,Membrane Glycoproteins ,biology ,MMP ,Uterus ,Trophoblast ,Placentation ,Cell Biology ,Cell biology ,Trophoblasts ,Pregnancy Trimester, First ,medicine.anatomical_structure ,Endocrinology ,Cell culture ,Giant cell ,EMILIN1 ,embryonic structures ,biology.protein ,Female ,Chorionic Villi ,Stromal Cells ,α4 integrin - Abstract
The detection of EMILIN1, a connective tissue glycoprotein associated with elastic fibers, at the level of the ectoplacental cone and trophoblast giant cells of developing mouse embryos (Braghetta et al., 2002) favored the idea of a structural as well as a functional role for this protein in the process of placentation. During the establishment of human placenta, a highly migratory subpopulation of extravillous trophoblasts (EVT), originating from anchoring chorionic villi, penetrate and invade the uterine wall. In this study we show that EMILIN1, produced by decidual stromal and smooth muscle uterine cells, is expressed in the stroma and in some instances as a gradient of increasing concentration in the perivascular region of modified vessels. This distribution pattern is consistent with the haptotactic directional migration observed in in vitro functional studies of freshly isolated EVT and of the immortalized HTR-8/SVneo cell line of trophoblasts. Function-blocking monoclonal antibodies against α4-integrin chain and against EMILIN1 as well as the use of EMILIN1-specific short interfering RNA confirmed that trophoblasts interact with EMILIN1 and/or its functional gC1q1 domain via α4β1 integrin. Finally, membrane type I-matrix metalloproteinase (MT1-MMP) and MMP-2 were upregulated in co-cultures of trophoblast cells and stromal cells, suggesting a contributing role in the haptotactic process towards EMILIN1.
- Published
- 2006
38. Abstract 3958: Multi-Reg: An integrative parallel approach to uncover drivers of cancer
- Author
-
Uri David Akavia, Carla Danussi, Felix Sanchez-Garcia, Antonio Iavarone, and Dana Pe'er
- Subjects
Cancer Research ,Oncology - Abstract
Tumor samples harbor a vast number of genomic alterations of various kinds, and it is difficult to distinguish driver alterations that contribute to oncogenesis from passenger alterations. Most computational methods identify driver genes by focusing on the most frequent copy number alterations. Previous work in our lab has led to the development of CONEXIC, a Bayesian algorithm that identifies candidate driver genes in cancer and links them to gene expression signatures they govern by integrating copy number and gene expression (Akavia et al, Cell, 2010). This algorithm was applied to data from melanoma cell lines, where it correctly identified known drivers, such as MITF and KLF6, and connected them to their known targets. In addition, it predicted novel tumor dependencies not previously implicated in melanoma, which were validated experimentally. Drivers may act concurrently, where not only the strongest one is important. For example, either PTEN deletion or AKT activation can lead to a similar expression signature and phenotype. Therefore, we developed a new algorithm, based on the same principles as CONEXIC, with multiple improvements. The new algorithm - Multi-Reg - is capable of detecting multiple candidate regulators that can all act in parallel to regulate an expression signature, and is also capable of integrating mutations in addition to copy number and gene expression data. We applied Multi-Reg to glioblastoma data from The Cancer Genome Atlas (TCGA). This data includes copy number, gene expression and mutations for hundreds of primary tumor samples. Multi-Reg has identified putative drivers that were missed by CONEXIC, such as the known genes PDGFRA and NF1, in addition to EGFR & ERB2 (identified by CONEXIC). Additionally, since Multi-Reg candidate drivers act in parallel, we can group them by shared targets. For example, EGFR & ERRB2 induce the same genes, which represent the Mesenchymal subtype of glibolastoma. These same genes are repressed by PDGFRA & NF1. This matches the known behavior of glioblastoma subtypes, where EGFR & PDGFRA characterize the Mesenchymal and Proneural subtypes, respectively. Thus, our results correctly identify known drivers of glioblastoma. Additionally, Multi-Reg results identified RHPN2 as a novel oncogenic factor controlling a gene expression signature related to adhesion. Validation has shown that while RHPN2 has no effect on cell proliferation it induces invasiveness in glioblastoma cell lines. This shows that Multi-Reg cannot only discover drivers, but can link them to the oncogenic phenomena they controls, and suggest the appropriate biological validation for it. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 3958. doi:1538-7445.AM2012-3958
- Published
- 2012
- Full Text
- View/download PDF
39. EMILIN1–α4/α9 integrin interaction inhibits dermal fibroblast and keratinocyte proliferation
- Author
-
Alessandra Petrucco, Bruna Wassermann, Paola Spessotto, Carla Danussi, Eliana Pivetta, Teresa Maria Elisa Modica, Lisa Del Bel Belluz, and Alfonso Colombatti
- Subjects
Keratinocytes ,Integrin alpha4 ,Integrin ,Extracellular matrix component ,Down-Regulation ,Smad2 Protein ,Integrin alpha4beta1 ,Corrections ,Article ,Dermal fibroblast ,Mice ,Phosphatidylinositol 3-Kinases ,medicine ,Animals ,Homeostasis ,Phosphorylation ,Cell adhesion ,Protein kinase B ,Research Articles ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Mice, Knockout ,Membrane Glycoproteins ,Mitogen-Activated Protein Kinase 3 ,Integrin alpha Chains ,integumentary system ,biology ,PTEN Phosphohydrolase ,Correction ,Dermis ,Cell Biology ,Fibroblasts ,Cell biology ,Enzyme Activation ,medicine.anatomical_structure ,biology.protein ,Keratinocyte ,Proto-Oncogene Proteins c-akt - Abstract
The α4/α9 integrins directly engage the ECM glycoprotein EMILIN1 to inhibit skin cell proliferation upstream of TGF-β signaling., EMILIN1 promotes α4β1 integrin–dependent cell adhesion and migration and reduces pro–transforming growth factor–β processing. A knockout mouse model was used to unravel EMILIN1 functions in skin where the protein was abundantly expressed in the dermal stroma and where EMILIN1-positive fibrils reached the basal keratinocyte layer. Loss of EMILIN1 caused dermal and epidermal hyperproliferation and accelerated wound closure. We identified the direct engagement of EMILIN1 to α4β1 and α9β1 integrins as the mechanism underlying the homeostatic role exerted by EMILIN1. The lack of EMILIN1–α4/α9 integrin interaction was accompanied by activation of PI3K/Akt and Erk1/2 pathways as a result of the reduction of PTEN. The down-regulation of PTEN empowered Erk1/2 phosphorylation that in turn inhibited Smad2 signaling by phosphorylation of residues Ser245/250/255. These results highlight the important regulatory role of an extracellular matrix component in skin proliferation. In addition, EMILIN1 is identified as a novel ligand for keratinocyte α9β1 integrin, suggesting prospective roles for this receptor–ligand pair in skin homeostasis.
- Published
- 2012
40. Abstract A26: Multi-Reg: An integrative parallel approach to uncover drivers of cancer
- Author
-
Dana Pe'er, Felix Sanchez-Garcia, Dylan Kotliar, Antonio Iavarone, Uri David Akavia, and Carla Danussi
- Subjects
Genetics ,Cancer Research ,biology ,PDGFRA ,medicine.disease ,medicine.disease_cause ,Microphthalmia-associated transcription factor ,Primary tumor ,Phenotype ,Oncology ,medicine ,biology.protein ,CEBPB ,PTEN ,Carcinogenesis ,Gene - Abstract
Tumor samples harbor a vast number of genomic alterations of various kinds, and it is not easy to distinguish driver alterations that contribute to oncogenesis from passenger alterations. Most computational methods attempt to identify driver alternations by focusing on the most frequent alterations. Previous work in our lab has led to the development of CONEXIC, a Bayesian framework for integrating copy number and gene expression to identify candidate driver genes in cancer and to link them to gene expression signatures they regulate (Akavia et al, Cell, 2010). This framework was applied to data from melanoma cell lines, where it correctly identified known drivers (MITF) and connected them to their known targets. In addition, it predicted novel tumor dependencies not previously implicated in melanoma, which were confirmed experimentally. In general, current algorithms identify only one driver (the strongest) controlling a gene expression signature. However, drivers may act in parallel, where not only the strongest one is important. For example, either PTEN deletion or AKT activation can lead to a similar expression signature and phenotype. Therefore, we developed a new algorithm, based on the same principles as CONEXIC with multiple improvements. The new algorithm (called Multi-Reg) is capable of detecting multiple candidate regulators that can all act in parallel to regulate an expression signature. This algorithm can integrate mutations in addition to copy number and expression. Finally, we have designed Multi-Reg to be easier and quicker to run and more robust. We applied Multi-Reg to glioblastoma data from The Cancer Genome Atlas (TCGA). This data includes copy number, gene expression and mutations for hundreds of primary tumor samples. We found 84 candidate regulators that were missed by CONEXIC, but discovered by Multi-Reg. Because of Multi-Reg's ability to search for genes working in parallel, it identified FGFR3, PDGFRA and NF1, in addition to EGFR & MET (identified by CONEXIC), as important candidate drivers. Additionally, Multi-Reg results identified RHPN2 as a novel oncogenic factor controlling a gene expression signature related to invasion and migration. Validation has shown that RHPN2 has limited effect on cell proliferation but induces invasiveness in glioblastoma cell lines. Our results correctly identify known drivers of glioblastoma progression, including oncogenes such as EGFR, MET, CEBPB and tumor suppressors such as p16 and NF1. Multi-Reg also has the capability to identify more regulators than CONEXIC, and has correctly linked RHPN2 to the oncogenic phenomena it controls. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the Second AACR International Conference on Frontiers in Basic Cancer Research; 2011 Sep 14-18; San Francisco, CA. Philadelphia (PA): AACR; Cancer Res 2011;71(18 Suppl):Abstract nr A26.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.