94 results on '"Massimo Squatrito"'
Search Results
2. A kinase-deficient NTRK2 splice variant predominates in glioma and amplifies several oncogenic signaling pathways
- Author
-
Siobhan S. Pattwell, Sonali Arora, Patrick J. Cimino, Tatsuya Ozawa, Frank Szulzewsky, Pia Hoellerbauer, Tobias Bonifert, Benjamin G. Hoffstrom, Norman E. Boiani, Hamid Bolouri, Colin E. Correnti, Barbara Oldrini, John R. Silber, Massimo Squatrito, Patrick J. Paddison, and Eric C. Holland
- Subjects
Science - Abstract
Tropomyosin receptor kinase B (TrkB), encoded by the neurotrophic tyrosine receptor kinase 2 (NTRK2) gene, exhibits intricate splicing patterns and post-translational modifications. Here, the authors perform whole gene and transcript-level analyses and report the TrkB.T1 splice variant enhances PDGF-driven gliomas in vivo and augments PI3K signaling cascades in vitro.
- Published
- 2020
- Full Text
- View/download PDF
3. Exploiting 4-1BB immune checkpoint to enhance the efficacy of oncolytic virotherapy for diffuse intrinsic pontine gliomas
- Author
-
Virginia Laspidea, Montserrat Puigdelloses, Sara Labiano, Lucía Marrodán, Marc Garcia-Moure, Marta Zalacain, Marisol Gonzalez-Huarriz, Naiara Martínez-Vélez, Iker Ausejo-Mauleon, Daniel de la Nava, Guillermo Herrador-Cañete, Javier Marco-Sanz, Elisabeth Guruceaga, Carlos E. de Andrea, María Villalba, Oren Becher, Massimo Squatrito, Verónica Matía, Jaime Gállego Pérez-Larraya, Ana Patiño-García, Sumit Gupta, Candelaria Gomez-Manzano, Juan Fueyo, and Marta M. Alonso
- Subjects
Immunology ,Oncology ,Medicine - Abstract
Diffuse intrinsic pontine gliomas (DIPGs) are aggressive pediatric brain tumors, and patient survival has not changed despite many therapeutic efforts, emphasizing the urgent need for effective treatments. Here, we evaluated the anti-DIPG effect of the oncolytic adenovirus Delta-24-ACT, which was engineered to express the costimulatory ligand 4-1BBL to potentiate the antitumor immune response of the virus. Delta-24-ACT induced the expression of functional 4-1BBL on the membranes of infected DIPG cells, which enhanced the costimulation of CD8+ T lymphocytes. In vivo, Delta-24-ACT treatment of murine DIPG orthotopic tumors significantly improved the survival of treated mice, leading to long-term survivors that developed immunological memory against these tumors. In addition, Delta-24-ACT was safe and caused no local or systemic toxicity. Mechanistic studies showed that Delta-24-ACT modulated the tumor-immune content, not only increasing the number, but also improving the functionality of immune cells. All of these data highlight the safety and potential therapeutic benefit of Delta-24-ACT the treatment of patients with DIPG.
- Published
- 2022
- Full Text
- View/download PDF
4. NF1 regulates mesenchymal glioblastoma plasticity and aggressiveness through the AP-1 transcription factor FOSL1
- Author
-
Carolina Marques, Thomas Unterkircher, Paula Kroon, Barbara Oldrini, Annalisa Izzo, Yuliia Dramaretska, Roberto Ferrarese, Eva Kling, Oliver Schnell, Sven Nelander, Erwin F Wagner, Latifa Bakiri, Gaetano Gargiulo, Maria Stella Carro, and Massimo Squatrito
- Subjects
GBM ,mesenchymal ,NF1 ,FOSL1 ,FRA-1 ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The molecular basis underlying glioblastoma (GBM) heterogeneity and plasticity is not fully understood. Using transcriptomic data of human patient-derived brain tumor stem cell lines (BTSCs), classified based on GBM-intrinsic signatures, we identify the AP-1 transcription factor FOSL1 as a key regulator of the mesenchymal (MES) subtype. We provide a mechanistic basis to the role of the neurofibromatosis type 1 gene (NF1), a negative regulator of the RAS/MAPK pathway, in GBM mesenchymal transformation through the modulation of FOSL1 expression. Depletion of FOSL1 in NF1-mutant human BTSCs and Kras-mutant mouse neural stem cells results in loss of the mesenchymal gene signature and reduction in stem cell properties and in vivo tumorigenic potential. Our data demonstrate that FOSL1 controls GBM plasticity and aggressiveness in response to NF1 alterations.
- Published
- 2021
- Full Text
- View/download PDF
5. LIF regulates CXCL9 in tumor-associated macrophages and prevents CD8+ T cell tumor-infiltration impairing anti-PD1 therapy
- Author
-
Mónica Pascual-García, Ester Bonfill-Teixidor, Ester Planas-Rigol, Carlota Rubio-Perez, Raffaella Iurlaro, Alexandra Arias, Isabel Cuartas, Ada Sala-Hojman, Laura Escudero, Francisco Martínez-Ricarte, Isabel Huber-Ruano, Paolo Nuciforo, Leire Pedrosa, Carolina Marques, Irene Braña, Elena Garralda, María Vieito, Massimo Squatrito, Estela Pineda, Francesc Graus, Carmen Espejo, Juan Sahuquillo, Josep Tabernero, and Joan Seoane
- Subjects
Science - Abstract
LIF is a pleiotropic cytokine that promotes an immunosuppressive microenvironment and has critical functions in embryonic development. Here, the authors show that LIF regulates CD8+ T cell tumor infiltration in cancer by repressing CXCL19 and promoting the presence of protumoral macrophages and thatLIF inhibition, via neutralizing antibodies, promotes T cell infiltration and synergizes with immune checkpoint inhbitors resulting in tumor regression and immunological memory.
- Published
- 2019
- Full Text
- View/download PDF
6. Somatic genome editing with the RCAS-TVA-CRISPR-Cas9 system for precision tumor modeling
- Author
-
Barbara Oldrini, Álvaro Curiel-García, Carolina Marques, Veronica Matia, Özge Uluçkan, Osvaldo Graña-Castro, Raul Torres-Ruiz, Sandra Rodriguez-Perales, Jason T. Huse, and Massimo Squatrito
- Subjects
Science - Abstract
Accurate recapitulation of human disease in animal models requires generation of complex and heterogeneous genetic variation. Here the authors combine RCAS-TVA with CRISPR-Cas9 to generate mouse models of cancer.
- Published
- 2018
- Full Text
- View/download PDF
7. EGFR feedback-inhibition by Ran-binding protein 6 is disrupted in cancer
- Author
-
Barbara Oldrini, Wan-Ying Hsieh, Hediye Erdjument-Bromage, Paolo Codega, Maria Stella Carro, Alvaro Curiel-García, Carl Campos, Maryam Pourmaleki, Christian Grommes, Igor Vivanco, Daniel Rohle, Craig M. Bielski, Barry S. Taylor, Travis J. Hollmann, Marc Rosenblum, Paul Tempst, John Blenis, Massimo Squatrito, and Ingo K. Mellinghoff
- Subjects
Science - Abstract
The epidermal growth factor receptor (EGFR) signalling is regulated at multiple levels. Here the authors show that the importin RanBP6 acts as a tumor suppressor in Glioblastoma and regulates EGFR signalling through promoting translocation of STAT3 to the nuclei and repressing EGFR transcription.
- Published
- 2017
- Full Text
- View/download PDF
8. Astrocyte-specific expression patterns associated with the PDGF-induced glioma microenvironment.
- Author
-
Amanda M Katz, Nduka M Amankulor, Ken Pitter, Karim Helmy, Massimo Squatrito, and Eric C Holland
- Subjects
Medicine ,Science - Abstract
The tumor microenvironment contains normal, non-neoplastic cells that may contribute to tumor growth and maintenance. Within PDGF-driven murine gliomas, tumor-associated astrocytes (TAAs) are a large component of the tumor microenvironment. The function of non-neoplastic astrocytes in the glioma microenvironment has not been fully elucidated; moreover, the differences between these astrocytes and normal astrocytes are unknown. We therefore sought to identify genes and pathways that are increased in TAAs relative to normal astrocytes and also to determine whether expression of these genes correlates with glioma behavior.We compared the gene expression profiles of TAAs to normal astrocytes and found the Antigen Presentation Pathway to be significantly increased in TAAs. We then identified a gene signature for glioblastoma (GBM) TAAs and validated the expression of some of those genes within the tumor. We also show that TAAs are derived from the non-tumor, stromal environment, in contrast to the Olig2+ tumor cells that constitute the neoplastic elements in our model. Finally, we validate this GBM TAA signature in patients and show that a TAA-derived gene signature predicts survival specifically in the human proneural subtype of glioma.Our data identifies unique gene expression patterns between populations of TAAs and suggests potential roles for stromal astrocytes within the glioma microenvironment. We show that certain stromal astrocytes in the tumor microenvironment express a GBM-specific gene signature and that the majority of these stromal astrocyte genes can predict survival in the human disease.
- Published
- 2012
- Full Text
- View/download PDF
9. Clonally expanded CD8 T cells characterize amyotrophic lateral sclerosis-4
- Author
-
Laura Campisi, Shahab Chizari, Jessica S. Y. Ho, Anastasia Gromova, Frederick J. Arnold, Lorena Mosca, Xueyan Mei, Yesai Fstkchyan, Denis Torre, Cindy Beharry, Marta Garcia-Forn, Miguel Jiménez-Alcázar, Vladislav A. Korobeynikov, Jack Prazich, Zahi A. Fayad, Marcus M. Seldin, Silvia De Rubeis, Craig L. Bennett, Lyle W. Ostrow, Christian Lunetta, Massimo Squatrito, Minji Byun, Neil A. Shneider, Ning Jiang, Albert R. La Spada, and Ivan Marazzi
- Subjects
Motor Neurons ,Multidisciplinary ,General Science & Technology ,Amyotrophic Lateral Sclerosis ,DNA Helicases ,Neurosciences ,CD8-Positive T-Lymphocytes ,Neurodegenerative ,Multifunctional Enzymes ,Article ,Clone Cells ,Brain Disorders ,Mice ,Rare Diseases ,Mutation ,Neurological ,Genetics ,Animals ,2.1 Biological and endogenous factors ,Gene Knock-In Techniques ,ALS ,Aetiology ,RNA Helicases - Abstract
Amyotrophic lateral sclerosis (ALS) is a heterogenous neurodegenerative disorder that affects motor neurons and voluntary muscle control1. ALS heterogeneity includes the age of manifestation, the rate of progression and the anatomical sites of symptom onset. Disease-causing mutations in specific genes have been identified and define different subtypes of ALS1. Although several ALS-associated genes have been shown to affect immune functions2, whether specific immune features account for ALS heterogeneity is poorly understood. Amyotrophic lateral sclerosis-4 (ALS4) is characterized by juvenile onset and slow progression3. Patients with ALS4 show motor difficulties by the time that they are in their thirties, and most of them require devices to assist with walking by their fifties. ALS4 is caused by mutations in the senataxin gene (SETX). Here, using Setx knock-in mice that carry the ALS4-causative L389S mutation, we describe an immunological signature that consists of clonally expanded, terminally differentiated effector memory (TEMRA) CD8 T cells in the central nervous system and the blood of knock-in mice. Increased frequencies of antigen-specific CD8 T cells in knock-in mice mirror the progression of motor neuron disease and correlate with anti-glioma immunity. Furthermore, bone marrow transplantation experiments indicate that the immune system has a key role in ALS4 neurodegeneration. In patients with ALS4, clonally expanded TEMRA CD8 T cells circulate in the peripheral blood. Our results provide evidence of an antigen-specific CD8 T cell response in ALS4, which could be used to unravel disease mechanisms and as a potential biomarker of disease state.
- Published
- 2022
- Full Text
- View/download PDF
10. Supplementary Data from Phenotypic Mapping of Pathologic Cross-Talk between Glioblastoma and Innate Immune Cells by Synthetic Genetic Tracing
- Author
-
Gaetano Gargiulo, Michela Serresi, Massimo Squatrito, Rainer Glass, Danielle Hulsman, Maria Pilar Sanchez-Bailon, Heike Naumann, Melanie Großmann, Sonia Kertalli, Andreas Göhrig, Iros Barozzi, Yuliia Dramaretska, Carlos Company, and Matthias Jürgen Schmitt
- Abstract
Supplementary Figures, legends and References
- Published
- 2023
- Full Text
- View/download PDF
11. Table S1 from Phenotypic Mapping of Pathologic Cross-Talk between Glioblastoma and Innate Immune Cells by Synthetic Genetic Tracing
- Author
-
Gaetano Gargiulo, Michela Serresi, Massimo Squatrito, Rainer Glass, Danielle Hulsman, Maria Pilar Sanchez-Bailon, Heike Naumann, Melanie Großmann, Sonia Kertalli, Andreas Göhrig, Iros Barozzi, Yuliia Dramaretska, Carlos Company, and Matthias Jürgen Schmitt
- Abstract
Supplementary Table S1. GBM-sLCR features
- Published
- 2023
- Full Text
- View/download PDF
12. Data from Dianhydrogalactitol Overcomes Multiple Temozolomide Resistance Mechanisms in Glioblastoma
- Author
-
Massimo Squatrito, Fátima Al-Shahrour, Scott W. Lowe, Lucía Zhu, Marcos Galán-Ganga, Alberto J. Schuhmacher, Javier Perales-Patón, Paula Nogales, Álvaro Curiel-García, and Miguel Jiménez-Alcázar
- Abstract
Glioblastoma (GBM) is the most frequent and aggressive primary tumor type in the central nervous system in adults. Resistance to chemotherapy remains one of the major obstacles in GBM treatment. Identifying and overcoming the mechanisms of therapy resistance is instrumental to develop novel therapeutic approaches for patients with GBM. To determine the major drivers of temozolomide (TMZ) sensitivity, we performed shRNA screenings in GBM lines with different O6-methylguanine-DNA methyl-transferase (MGMT) status. We then evaluated dianhydrogalactitol (Val-083), a small alkylating molecule that induces interstrand DNA crosslinking, as a potential treatment to bypass TMZ-resistance mechanisms. We found that loss of mismatch repair (MMR) components and MGMT expression are mutually exclusive mechanisms driving TMZ resistance in vitro. Treatment of established GBM cells and tumorsphere lines with Val-083 induces DNA damage and cell-cycle arrest in G2–M phase, independently of MGMT or MMR status, thus circumventing conventional resistance mechanisms to TMZ. Combination of TMZ and Val-083 shows a synergic cytotoxic effect in tumor cells in vitro, ex vivo, and in vivo. We propose this combinatorial treatment as a potential approach for patients with GBM.
- Published
- 2023
- Full Text
- View/download PDF
13. Table S2 from Dianhydrogalactitol Overcomes Multiple Temozolomide Resistance Mechanisms in Glioblastoma
- Author
-
Massimo Squatrito, Fátima Al-Shahrour, Scott W. Lowe, Lucía Zhu, Marcos Galán-Ganga, Alberto J. Schuhmacher, Javier Perales-Patón, Paula Nogales, Álvaro Curiel-García, and Miguel Jiménez-Alcázar
- Abstract
Screening results U251 TMZ
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Figures from Dianhydrogalactitol Overcomes Multiple Temozolomide Resistance Mechanisms in Glioblastoma
- Author
-
Massimo Squatrito, Fátima Al-Shahrour, Scott W. Lowe, Lucía Zhu, Marcos Galán-Ganga, Alberto J. Schuhmacher, Javier Perales-Patón, Paula Nogales, Álvaro Curiel-García, and Miguel Jiménez-Alcázar
- Abstract
Combined supplementary Figures
- Published
- 2023
- Full Text
- View/download PDF
15. Data from Phenotypic Mapping of Pathologic Cross-Talk between Glioblastoma and Innate Immune Cells by Synthetic Genetic Tracing
- Author
-
Gaetano Gargiulo, Michela Serresi, Massimo Squatrito, Rainer Glass, Danielle Hulsman, Maria Pilar Sanchez-Bailon, Heike Naumann, Melanie Großmann, Sonia Kertalli, Andreas Göhrig, Iros Barozzi, Yuliia Dramaretska, Carlos Company, and Matthias Jürgen Schmitt
- Abstract
Glioblastoma is a lethal brain tumor that exhibits heterogeneity and resistance to therapy. Our understanding of tumor homeostasis is limited by a lack of genetic tools to selectively identify tumor states and fate transitions. Here, we use glioblastoma subtype signatures to construct synthetic genetic tracing cassettes and investigate tumor heterogeneity at cellular and molecular levels, in vitro and in vivo. Through synthetic locus control regions, we demonstrate that proneural glioblastoma is a hardwired identity, whereas mesenchymal glioblastoma is an adaptive and metastable cell state driven by proinflammatory and differentiation cues and DNA damage, but not hypoxia. Importantly, we discovered that innate immune cells divert glioblastoma cells to a proneural-to-mesenchymal transition that confers therapeutic resistance. Our synthetic genetic tracing methodology is simple, scalable, and widely applicable to study homeostasis in development and diseases. In glioblastoma, the method causally links distinct (micro)environmental, genetic, and pharmacologic perturbations and mesenchymal commitment.Significance:Glioblastoma is heterogeneous and incurable. Here, we designed synthetic reporters to reflect the transcriptional output of tumor cell states and signaling pathways' activity. This method is generally applicable to study homeostasis in normal tissues and diseases. In glioblastoma, synthetic genetic tracing causally connects cellular and molecular heterogeneity to therapeutic responses.This article is highlighted in the In This Issue feature, p. 521
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Data from The Irradiated Brain Microenvironment Supports Glioma Stemness and Survival via Astrocyte-Derived Transglutaminase 2
- Author
-
Alexander Pietras, Massimo Squatrito, Håkan Axelson, Mattias Belting, Johan Bengzon, Valeria Governa, Fredrik J. Swartling, Tobias Bergström, Elin J. Pietras, Pauline Jeannot, David Lindgren, Kristoffer von Stedingk, Elinn Johansson, Vasiliki Pantazopoulou, Carolina Marques, and Tracy J. Berg
- Abstract
Supplementary Tables
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Figures from The Irradiated Brain Microenvironment Supports Glioma Stemness and Survival via Astrocyte-Derived Transglutaminase 2
- Author
-
Alexander Pietras, Massimo Squatrito, Håkan Axelson, Mattias Belting, Johan Bengzon, Valeria Governa, Fredrik J. Swartling, Tobias Bergström, Elin J. Pietras, Pauline Jeannot, David Lindgren, Kristoffer von Stedingk, Elinn Johansson, Vasiliki Pantazopoulou, Carolina Marques, and Tracy J. Berg
- Abstract
Supplementary Figures 1-6
- Published
- 2023
- Full Text
- View/download PDF
18. Data from The Irradiated Brain Microenvironment Supports Glioma Stemness and Survival via Astrocyte-Derived Transglutaminase 2
- Author
-
Alexander Pietras, Massimo Squatrito, Håkan Axelson, Mattias Belting, Johan Bengzon, Valeria Governa, Fredrik J. Swartling, Tobias Bergström, Elin J. Pietras, Pauline Jeannot, David Lindgren, Kristoffer von Stedingk, Elinn Johansson, Vasiliki Pantazopoulou, Carolina Marques, and Tracy J. Berg
- Abstract
The tumor microenvironment plays an essential role in supporting glioma stemness and radioresistance. Following radiotherapy, recurrent gliomas form in an irradiated microenvironment. Here we report that astrocytes, when pre-irradiated, increase stemness and survival of cocultured glioma cells. Tumor-naïve brains increased reactive astrocytes in response to radiation, and mice subjected to radiation prior to implantation of glioma cells developed more aggressive tumors. Extracellular matrix derived from irradiated astrocytes were found to be a major driver of this phenotype and astrocyte-derived transglutaminase 2 (TGM2) was identified as a promoter of glioma stemness and radioresistance. TGM2 levels increased after radiation in vivo and in recurrent human glioma, and TGM2 inhibitors abrogated glioma stemness and survival. These data suggest that irradiation of the brain results in the formation of a tumor-supportive microenvironment. Therapeutic targeting of radiation-induced, astrocyte-derived extracellular matrix proteins may enhance the efficacy of standard-of-care radiotherapy by reducing stemness in glioma.Significance:These findings presented here indicate that radiotherapy can result in a tumor-supportive microenvironment, the targeting of which may be necessary to overcome tumor cell therapeutic resistance and recurrence.
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Methods from The Irradiated Brain Microenvironment Supports Glioma Stemness and Survival via Astrocyte-Derived Transglutaminase 2
- Author
-
Alexander Pietras, Massimo Squatrito, Håkan Axelson, Mattias Belting, Johan Bengzon, Valeria Governa, Fredrik J. Swartling, Tobias Bergström, Elin J. Pietras, Pauline Jeannot, David Lindgren, Kristoffer von Stedingk, Elinn Johansson, Vasiliki Pantazopoulou, Carolina Marques, and Tracy J. Berg
- Abstract
Extended experimental details to supplement the main article file methods section
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Table 1 from 53BP1 Is a Haploinsufficient Tumor Suppressor and Protects Cells from Radiation Response in Glioma
- Author
-
Eric C. Holland, Maria Jasin, Nikolaus Schultz, Fabio Vanoli, and Massimo Squatrito
- Abstract
XLS file - 102K, TCGA data for TP53BP1 in the GBM dataset
- Published
- 2023
- Full Text
- View/download PDF
21. Supplementary Methods from 53BP1 Is a Haploinsufficient Tumor Suppressor and Protects Cells from Radiation Response in Glioma
- Author
-
Eric C. Holland, Maria Jasin, Nikolaus Schultz, Fabio Vanoli, and Massimo Squatrito
- Abstract
PDF file - 132K
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Figure 4 from 53BP1 Is a Haploinsufficient Tumor Suppressor and Protects Cells from Radiation Response in Glioma
- Author
-
Eric C. Holland, Maria Jasin, Nikolaus Schultz, Fabio Vanoli, and Massimo Squatrito
- Abstract
PDF file - 130K, Cell cycle alteration in 53BP1 silenced cells, upon exposure to IR
- Published
- 2023
- Full Text
- View/download PDF
23. Supplementary Figure 2 from 53BP1 Is a Haploinsufficient Tumor Suppressor and Protects Cells from Radiation Response in Glioma
- Author
-
Eric C. Holland, Maria Jasin, Nikolaus Schultz, Fabio Vanoli, and Massimo Squatrito
- Abstract
PDF file - 310K, 53BP1 null mouse cells, but not heterozygous, and robust silencing in GBM cells lead to high sensitivity to IR
- Published
- 2023
- Full Text
- View/download PDF
24. Supplementary Table 2 from 53BP1 Is a Haploinsufficient Tumor Suppressor and Protects Cells from Radiation Response in Glioma
- Author
-
Eric C. Holland, Maria Jasin, Nikolaus Schultz, Fabio Vanoli, and Massimo Squatrito
- Abstract
XLS file - 66K, Co-occurrence of TP53BP1 and TP53 alterations in the TCGA dataset for GBM
- Published
- 2023
- Full Text
- View/download PDF
25. Supplementary Figure 5 from 53BP1 Is a Haploinsufficient Tumor Suppressor and Protects Cells from Radiation Response in Glioma
- Author
-
Eric C. Holland, Maria Jasin, Nikolaus Schultz, Fabio Vanoli, and Massimo Squatrito
- Abstract
PDF file - 205, 53BP1 modulates NHEJ in glioma cell lines
- Published
- 2023
- Full Text
- View/download PDF
26. Supplementary Figure 3 from 53BP1 Is a Haploinsufficient Tumor Suppressor and Protects Cells from Radiation Response in Glioma
- Author
-
Eric C. Holland, Maria Jasin, Nikolaus Schultz, Fabio Vanoli, and Massimo Squatrito
- Abstract
PDF file - 22K, 53BP1 silencing does not increase TMZ sensitivity in T98G cells
- Published
- 2023
- Full Text
- View/download PDF
27. Supplementary Materials and Methods and Figures from Histone Acetyltransferase PCAF Is Required for Hedgehog–Gli-Dependent Transcription and Cancer Cell Proliferation
- Author
-
Kristian Helin, Massimo Squatrito, Deo P. Pandey, Faizaan Mohammad, Cornelia Steinhauer, and Martina Malatesta
- Abstract
PDF file, 906K, Supplementary Materials and Methods Flow cytometry. Colony formation assay. Supplementary Tables Supplementary Table 1. siRNAs and shRNAs constructs used in this study. Supplementary Table 2. Expression primers and ChIP primers used in this study. Supplementary Table 3. Antibodies used in this study. Supplementary Figures Supplementary Figure S1. Analysis of siRNA screening for acetyltransferases. Supplementary Figure S2. Validation of candidate genes from the siRNA screening. Supplementary Figure S3. Stable depletion of Pcaf does not affect DNA synthesis in NIH3T3 cells. Supplementary Figure S4. Inhibition of PCAF expression affects neurosphere formation and proliferation of GBM543 cells. Supplementary Figure S5. Depletion of PCAF and inhibitors to PCAF induce apoptosis. Supplementary Figure S6. Inhibition of Pcaf expression affects proliferation of tNSCs.
- Published
- 2023
- Full Text
- View/download PDF
28. Dianhydrogalactitol Overcomes Multiple Temozolomide Resistance Mechanisms in Glioblastoma
- Author
-
Marcos Galán-Ganga, Alberto J. Schuhmacher, Paula Nogales, Fatima Al-Shahrour, Miguel Jiménez-Alcázar, Lucía Zhu, Javier Perales-Patón, Scott W. Lowe, Álvaro Curiel-García, and Massimo Squatrito
- Subjects
0301 basic medicine ,Cancer Research ,DNA damage ,medicine.medical_treatment ,Transfection ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Temozolomide ,medicine ,Animals ,Humans ,Dianhydrogalactitol ,Chemotherapy ,business.industry ,medicine.disease ,Xenograft Model Antitumor Assays ,Primary tumor ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,DNA mismatch repair ,Glioblastoma ,business ,Ex vivo ,medicine.drug - Abstract
Glioblastoma (GBM) is the most frequent and aggressive primary tumor type in the central nervous system in adults. Resistance to chemotherapy remains one of the major obstacles in GBM treatment. Identifying and overcoming the mechanisms of therapy resistance is instrumental to develop novel therapeutic approaches for patients with GBM. To determine the major drivers of temozolomide (TMZ) sensitivity, we performed shRNA screenings in GBM lines with different O6-methylguanine-DNA methyl-transferase (MGMT) status. We then evaluated dianhydrogalactitol (Val-083), a small alkylating molecule that induces interstrand DNA crosslinking, as a potential treatment to bypass TMZ-resistance mechanisms. We found that loss of mismatch repair (MMR) components and MGMT expression are mutually exclusive mechanisms driving TMZ resistance in vitro. Treatment of established GBM cells and tumorsphere lines with Val-083 induces DNA damage and cell-cycle arrest in G2–M phase, independently of MGMT or MMR status, thus circumventing conventional resistance mechanisms to TMZ. Combination of TMZ and Val-083 shows a synergic cytotoxic effect in tumor cells in vitro, ex vivo, and in vivo. We propose this combinatorial treatment as a potential approach for patients with GBM.
- Published
- 2021
- Full Text
- View/download PDF
29. The Irradiated Brain Microenvironment Supports Glioma Stemness and Survival via Astrocyte-Derived Transglutaminase 2
- Author
-
Pauline Jeannot, Fredrik J. Swartling, Elinn Johansson, David Lindgren, Carolina Marques, Tracy J. Berg, Håkan Axelson, Mattias Belting, Johan Bengzon, Massimo Squatrito, Kristoffer von Stedingk, Alexander Pietras, Elin J. Pietras, Vasiliki Pantazopoulou, Tobias Bergström, and Valeria Governa
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Cell Survival ,Tissue transglutaminase ,medicine.medical_treatment ,Radiation Tolerance ,Extracellular matrix ,Mice ,03 medical and health sciences ,0302 clinical medicine ,GTP-Binding Proteins ,In vivo ,Glioma ,Radioresistance ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Protein Glutamine gamma Glutamyltransferase 2 ,Enzyme Inhibitors ,Tumor microenvironment ,Transglutaminases ,biology ,Brain Neoplasms ,Chemistry ,Brain ,medicine.disease ,Extracellular Matrix ,Radiation therapy ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Astrocytes ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Cancer research ,biology.protein ,Female ,Neoplasm Recurrence, Local ,Glioblastoma ,Astrocyte - Abstract
The tumor microenvironment plays an essential role in supporting glioma stemness and radioresistance. Following radiotherapy, recurrent gliomas form in an irradiated microenvironment. Here we report that astrocytes, when pre-irradiated, increase stemness and survival of cocultured glioma cells. Tumor-naïve brains increased reactive astrocytes in response to radiation, and mice subjected to radiation prior to implantation of glioma cells developed more aggressive tumors. Extracellular matrix derived from irradiated astrocytes were found to be a major driver of this phenotype and astrocyte-derived transglutaminase 2 (TGM2) was identified as a promoter of glioma stemness and radioresistance. TGM2 levels increased after radiation in vivo and in recurrent human glioma, and TGM2 inhibitors abrogated glioma stemness and survival. These data suggest that irradiation of the brain results in the formation of a tumor-supportive microenvironment. Therapeutic targeting of radiation-induced, astrocyte-derived extracellular matrix proteins may enhance the efficacy of standard-of-care radiotherapy by reducing stemness in glioma. Significance: These findings presented here indicate that radiotherapy can result in a tumor-supportive microenvironment, the targeting of which may be necessary to overcome tumor cell therapeutic resistance and recurrence.
- Published
- 2021
- Full Text
- View/download PDF
30. CTNI-03. A PHASE I/IIA, OPEN-LABEL STUDY OF THE BRAIN-PENETRANT PARP1-SELECTIVE INHIBITOR AZD9574 AS MONOTHERAPY AND IN COMBINATION IN PATIENTS WITH ADVANCED SOLID MALIGNANCIES (CERTIS1)
- Author
-
Nancy Mueller, Stephen Luen, Roger Stupp, Anthony Chalmers, Baisong Huang, Massimo Squatrito, Barry Davies, Petra Hamerlik, and Timothy Yap
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
BACKGROUND Currently approved Poly ADP-Ribose Polymerase (PARP) inhibitors (PARPi) selectively inhibit and trap both PARP1 and PARP2 (PARP1/2) at sites of single strand (ss) deoxyribonucleic acid (DNA) (ssDNA) damage, preventing ssDNA repair and leading to replication-dependent DNA double strand breaks. Recent data suggest that only inhibition of PARP1 is required for anti-proliferative effects, while PARP2 functions in the survival of haematopoietic stem and progenitor cells. These observations suggest that the inhibition and trapping of PARP 2 is not needed for anti-cancer effects, and may be a major driver of haematological toxicity observed. AZD9574 is a novel brain-penetrant PARPi that potently and selectively inhibits and traps PARP1, with the goal of delivering efficacious, less toxic, and more combinable PARPi. Furthermore, owing to its central nervous system penetration capability, AZD9574 may provide a new treatment option for patients with CNS malignancies or patients with brain metastases characterized by homologous recombination deficiency (HRD). METHODS This is a first-in-human modular study primarily designed to evaluate the safety and tolerability of AZD9574 as monotherapy and in combination with anti-cancer agents at increasing dose levels in patients with advanced solid malignancies, followed by expansion cohorts in specific indications. The study will also characterize the pharmacokinetics of AZD9574 and explore potential biological activity by assessing pharmacodynamic and exploratory biomarkers and anti-tumour activity. Module 1 will enrol patients with advanced breast, ovarian, pancreatic or prostate tumours harbouring homologous recombination deficiencies. Module 2 will enrol patients with isocitrate dehydrogenase (IDH)1/2 mutated glioma.
- Published
- 2022
- Full Text
- View/download PDF
31. Author Correction: Clonally expanded CD8 T cells characterize amyotrophic lateral sclerosis-4
- Author
-
Laura Campisi, Shahab Chizari, Jessica S. Y. Ho, Anastasia Gromova, Frederick J. Arnold, Lorena Mosca, Xueyan Mei, Yesai Fstkchyan, Denis Torre, Cindy Beharry, Marta Garcia-Forn, Miguel Jiménez-Alcázar, Vladislav A. Korobeynikov, Jack Prazich, Zahi A. Fayad, Marcus M. Seldin, Silvia De Rubeis, Craig L. Bennett, Lyle W. Ostrow, Christian Lunetta, Massimo Squatrito, Minji Byun, Neil A. Shneider, Ning Jiang, Albert R. La Spada, and Ivan Marazzi
- Subjects
Multidisciplinary - Published
- 2022
- Full Text
- View/download PDF
32. Author response: NF1 regulates mesenchymal glioblastoma plasticity and aggressiveness through the AP-1 transcription factor FOSL1
- Author
-
Carolina Marques, Roberto Ferrarese, Gaetano Gargiulo, Yuliia Dramaretska, Maria Stella Carro, Erwin F. Wagner, Massimo Squatrito, Latifa Bakiri, Paula Kroon, Eva Kling, Annalisa Izzo, Oliver Schnell, Barbara Oldrini, Thomas Unterkircher, and Sven Nelander
- Subjects
AP-1 transcription factor ,Mesenchymal Glioblastoma ,Cancer research ,Plasticity ,Biology ,FOSL1 - Published
- 2021
- Full Text
- View/download PDF
33. Abstract 1331: Elucidating cell-to-cell communication and immunotherapy responses in deeply characterized mouse models of human glioma subtypes
- Author
-
Caiyi Wang, Jose Maldonado, Thomas D. Gallup, Nourhan Abdelfattah, Jia-Shiun Leu, Nithin Joshy, joshy George, Jihye Paik, Massimo Squatrito, and Kyuson Yun
- Subjects
Cancer Research ,Oncology - Abstract
Immunotherapy is a promising treatment modality for highly invasive gliomas; however, clinical trials thus far have failed to provide significant clinical benefit to most GBM patients. GBM is one of the “immune cold” tumors characterized by poor infiltration of T cells even though greater than 40% of glioma cells are composed of immune cells in some patients. The majority of immune cells in GBM are immune-suppressive myeloid cells that block T cell infiltration and/or activation. To elucidate the mechanisms of immune evasion and to understand how the immune system interacts with glioma and stromal cells that shape the immune-suppressive landscape in GBM, there is an urgent need for immune-competent preclinical models that recapitulate the human disease. Human GBM is divided into three molecular subtypes (proneural:PR, classical:CL, and mesenchymal:MES) based on specific gene expression patterns and signature mutational profiles. Here, we report multi-dimensional analyses of six different transplantable mouse glioma models in the C57BL6/J background that represent all three human GBM molecular subtypes. We performed whole-exome sequencing as well as STR fingerprinting of each primary tumorsphere line and also performed immune phenotyping of each glioma model with flow cytometry. To gain molecular insights and determine cellular heterogeneity, we also performed single-cell RNA sequencing from the six models. Glioma cell analysis at the single-cell level revealed that cell-of-origin rather than the oncogenic driver (such as EGFRviii) plays a dominant role in determining the molecular phenotypes of glioma cells, driving their classification into a molecular subtype defined by human studies. In addition, we identified eight molecular subtypes of glioma-associated myeloid (GAMs), seven different subtypes of T cells, and provide molecular definitions of glioma-associated pericytes and endothelial cells in murine gliomas. In addition, we performed cross-species comparisons of glioma and immune cell subtypes between humans and mice at the single-cell level. Furthermore, we report qualitative and quantitative differences in the cell-to-cell communication among different stromal cells and glioma cells in each model, and propose that these interactions shape the local niche and functional neighborhoods. Finally, we leverage these preclinical models to elucidate underlying molecular mechanisms that drive differential sensitivities of each model to immunotherapies: anti-PD1, CTLA4, and 4-1BB in vivo. In summary, we report deeply characterized mouse models of human GBM subtypes and highlight their utility as preclinical models for immunotherapy evaluation and foundational tumor immunology studies. Citation Format: Caiyi Wang, Jose Maldonado, Thomas D. Gallup, Nourhan Abdelfattah, Jia-Shiun Leu, Nithin Joshy, joshy George, Jihye Paik, Massimo Squatrito, Kyuson Yun. Elucidating cell-to-cell communication and immunotherapy responses in deeply characterized mouse models of human glioma subtypes [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 1331.
- Published
- 2022
- Full Text
- View/download PDF
34. Identification of a dexamethasone mediated radioprotection mechanism reveals new therapeutic vulnerabilities in glioblastoma
- Author
-
Estefania Carrasco-Garcia, Irene Lasheras-Otero, Larraitz Egaña, Imanol Arozarena, Marta Redondo-Muñoz, Iñaki M Larráyoz, Marta Maria-Alonso, Massimo Squatrito, Paula Aldaz, Joaquín Fernández-Irigoyen, Nicolás Samprón, Idoia Morilla, M Victoria Zelaya, Claudia Wellbrock, Jaione Auzmendi-Iriarte, Ana Olías-Arjona, Enrique Santamaría, Ander Matheu, Mikel Rico, Laura Bragado, and Maika Durantez
- Subjects
musculoskeletal diseases ,0301 basic medicine ,mitosis checkpoint ,Cancer Research ,PDGFR ,sunitinib ,Cell ,dexamethasone ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,Glioma ,medicine ,biology ,Sunitinib ,business.industry ,Ponatinib ,glioblastoma ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,musculoskeletal system ,medicine.disease ,Dasatinib ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,business ,Tyrosine kinase ,Platelet-derived growth factor receptor ,medicine.drug - Abstract
Simple Summary The standard of care for patients with newly diagnosed glioblastoma (GBM) comprises surgery followed by radio- and chemotherapy. In addition, dexamethasone is used to manage the development of inflammation within the brain in general, and particularly during treatment. The effects of dexamethasone on patient survival however remain controversial because several clinical studies suggest that dexamethasone could potentially restrict effective radiotherapy. With the idea to improve GBM therapy, we set out to identify small molecule inhibitors that could improve the killing of GBM cells when applied together with radiotherapy. We have identified a novel dexamethasone-induced mechanism that can directly protect GBM cells from radiotherapy and thus may contribute to the adverse effects observed in the clinic. Strikingly, this mechanism also sensitises GBM cells to tyrosine kinase inhibitors, thus encouraging the revision of the use of these inhibitors for the treatment of GBM, potentially in an adjuvant setting. Abstract (1) Background: Despite the indisputable effectiveness of dexamethasone (DEXA) to reduce inflammation in glioblastoma (GBM) patients, its influence on tumour progression and radiotherapy response remains controversial. (2) Methods: We analysed patient data and used expression and cell biological analyses to assess effects of DEXA on GBM cells. We tested the efficacy of tyrosine kinase inhibitors in vitro and in vivo. (3) Results: We confirm in our patient cohort that administration of DEXA correlates with worse overall survival and shorter time to relapse. In GBM cells and glioma stem-like cells (GSCs) DEXA down-regulates genes controlling G2/M and mitotic-spindle checkpoints, and it enables cells to override the spindle assembly checkpoint (SAC). Concurrently, DEXA up-regulates Platelet Derived Growth Factor Receptor (PDGFR) signalling, which stimulates expression of anti-apoptotic regulators BCL2L1 and MCL1, required for survival during extended mitosis. Importantly, the protective potential of DEXA is dependent on intact tyrosine kinase signalling and ponatinib, sunitinib and dasatinib, all effectively overcome the radio-protective and pro-proliferative activity of DEXA. Moreover, we discovered that DEXA-induced signalling creates a therapeutic vulnerability for sunitinib in GSCs and GBM cells in vitro and in vivo. (4) Conclusions: Our results reveal a novel DEXA-induced mechanism in GBM cells and provide a rationale for revisiting the use of tyrosine kinase inhibitors for the treatment of GBM.
- Published
- 2021
35. NF1 regulates mesenchymal gliblastoma plasticity and aggressiveness through the AP-1 transcription factor FOSL1
- Author
-
Latifa Bakiri, Barbara Oldrini, Eva Kling, Roberto Ferrarese, Gaetano Gargiulo, Massimo Squatrito, Thomas Unterkircher, Annalisa Izzo, Sven Nelander, Maria Stella Carro, Paula Kroon, Oliver Schnell, Yuliia Dramaretska, Erwin F. Wagner, and Carolina Marques
- Subjects
Cancer Research ,Mouse ,QH301-705.5 ,Science ,Mesenchymal Glioblastoma ,Regulator ,Medical Biotechnology (with a focus on Cell Biology (including Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy) ,Biology ,mesenchymal ,FOSL1 ,GBM ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Humans ,Biology (General) ,Transcription factor ,Medicinsk bioteknologi (med inriktning mot cellbiologi (inklusive stamcellsbiologi), molekylärbiologi, mikrobiologi, biokemi eller biofarmaci) ,FRA-1 ,030304 developmental biology ,Cancer Biology ,0303 health sciences ,Neurofibromin 1 ,General Immunology and Microbiology ,Brain Neoplasms ,General Neuroscience ,Mesenchymal stem cell ,General Medicine ,Gene signature ,Neural stem cell ,nervous system diseases ,Gene Expression Regulation, Neoplastic ,AP-1 transcription factor ,NF1 ,030220 oncology & carcinogenesis ,Cancer research ,Neoplastic Stem Cells ,Medicine ,Stem cell ,Glioblastoma ,Proto-Oncogene Proteins c-fos ,Research Article ,Human - Abstract
The molecular basis underlying glioblastoma (GBM) heterogeneity and plasticity is not fully understood. Using transcriptomic data of human patient-derived brain tumor stem cell lines (BTSCs), classified based on GBM-intrinsic signatures, we identify the AP-1 transcription factor FOSL1 as a key regulator of the mesenchymal (MES) subtype. We provide a mechanistic basis to the role of the neurofibromatosis type 1 gene (NF1), a negative regulator of the RAS/MAPK pathway, in GBM mesenchymal transformation through the modulation of FOSL1 expression. Depletion of FOSL1 in NF1-mutant human BTSCs and Kras-mutant mouse neural stem cells results in loss of the mesenchymal gene signature and reduction in stem cell properties and in vivo tumorigenic potential. Our data demonstrate that FOSL1 controls GBM plasticity and aggressiveness in response to NF1 alterations.
- Published
- 2021
36. A kinase-deficient NTRK2 splice variant predominates in glioma and amplifies several oncogenic signaling pathways
- Author
-
Patrick J. Cimino, Sonali Arora, Tobias Bonifert, Norman Boiani, Tatsuya Ozawa, Barbara Oldrini, Frank Szulzewsky, Eric C. Holland, Hamid Bolouri, Massimo Squatrito, Siobhan S. Pattwell, Colin Correnti, Patrick J. Paddison, Pia Hoellerbauer, Benjamin G. Hoffstrom, John R. Silber, United States Department of Health & Human Services National Institutes of Health (NIH) - USA, American Cancer Society, French National Research Agency (ANR), Alzheimer's Disease Research Center, United States of Department of Health & Human Services, and French Development Agency
- Subjects
0301 basic medicine ,RNA splicing ,Oncogene Proteins, Fusion ,Carcinogenesis ,RNA Splicing ,Science ,General Physics and Astronomy ,Tropomyosin receptor kinase B ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Fusion gene ,03 medical and health sciences ,Mice ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Neural Stem Cells ,RNA Isoforms ,Animals ,Humans ,Receptor, trkB ,splice ,lcsh:Science ,Protein kinase B ,Cells, Cultured ,Multidisciplinary ,Membrane Glycoproteins ,Brain Neoplasms ,Gene Expression Profiling ,Alternative splicing ,Brain ,High-Throughput Nucleotide Sequencing ,Neurotrophic factors ,General Chemistry ,Glioma ,Oncogenes ,Cell biology ,Gene expression profiling ,CNS cancer ,030104 developmental biology ,Gene Ontology ,nervous system ,030220 oncology & carcinogenesis ,NIH 3T3 Cells ,lcsh:Q ,Signal transduction ,Signal Transduction - Abstract
Independent scientific achievements have led to the discovery of aberrant splicing patterns in oncogenesis, while more recent advances have uncovered novel gene fusions involving neurotrophic tyrosine receptor kinases (NTRKs) in gliomas. The exploration of NTRK splice variants in normal and neoplastic brain provides an intersection of these two rapidly evolving fields. Tropomyosin receptor kinase B (TrkB), encoded NTRK2, is known for critical roles in neuronal survival, differentiation, molecular properties associated with memory, and exhibits intricate splicing patterns and post-translational modifications. Here, we show a role for a truncated NTRK2 splice variant, TrkB.T1, in human glioma. TrkB.T1 enhances PDGF-driven gliomas in vivo, augments PDGF-induced Akt and STAT3 signaling in vitro, while next generation sequencing broadly implicates TrkB.T1 in the PI3K signaling cascades in a ligand-independent fashion. These TrkB.T1 findings highlight the importance of expanding upon whole gene and gene fusion analyses to include splice variants in basic and translational neuro-oncology research., Tropomyosin receptor kinase B (TrkB), encoded by the neurotrophic tyrosine receptor kinase 2 (NTRK2) gene, exhibits intricate splicing patterns and post-translational modifications. Here, the authors perform whole gene and transcript-level analyses and report the TrkB.T1 splice variant enhances PDGF-driven gliomas in vivo and augments PI3K signaling cascades in vitro.
- Published
- 2020
37. The mechanistic GEMMs of oncogenic histones
- Author
-
Massimo Squatrito, Anders Lindroth, Yoon Jung Park, and Veronica Matia
- Subjects
Biology ,medicine.disease_cause ,Epigenesis, Genetic ,Histones ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Age groups ,Neoplasms ,Genetics ,medicine ,Nucleosome ,Animals ,Humans ,Molecular Biology ,Genetics (clinical) ,030304 developmental biology ,0303 health sciences ,Biological modeling ,General Medicine ,Histone-Lysine N-Methyltransferase ,Tumor formation ,Nucleosomes ,Gene Expression Regulation, Neoplastic ,Crosstalk (biology) ,Disease Models, Animal ,Histone ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,biology.protein ,Carcinogenesis - Abstract
The last decade’s progress unraveling the mutational landscape of all age groups of cancer has uncovered mutations in histones as vital contributors of tumorigenesis. Here we review three new aspects of oncogenic histones: first, the identification of additional histone mutations potentially contributing to cancer formation; second, tumors expressing histone mutations to study the crosstalk of post-translational modifications, and; third, development of sophisticated biological model systems to reproduce tumorigenesis. At the outset, we recapitulate the firstly discovered histone mutations in pediatric and adolescent tumors of the brain and bone, which still remain the most pronounced histone alterations in cancer. We branch out to discuss the ramifications of histone mutations, including novel ones, that stem from altered protein-protein interactions of cognate histone modifiers as well as the stability of the nucleosome. We close by discussing animal models of oncogenic histones that reproduce tumor formation molecularly and morphologically and the prospect of utilizing them for drug testing, leading to efficient treatment and cure of deadly cancers with histone mutations.
- Published
- 2020
38. The irradiated brain microenvironment supports glioma stemness and survival via astrocyte-derived Transglutaminase 2
- Author
-
Tracy J. Berg, Carolina Marques, Vasiliki Pantazopoulou, Elinn Johansson, Kristoffer von Stedingk, David Lindgren, Elin J. Pietras, Tobias Bergström, Fredrik J. Swartling, Valeria Governa, Johan Bengzon, Mattias Belting, Håkan Axelson, Massimo Squatrito, and Alexander Pietras
- Subjects
Tumor microenvironment ,biology ,Tissue transglutaminase ,Chemistry ,medicine.disease ,In vitro ,Extracellular matrix ,medicine.anatomical_structure ,Radioresistance ,Glioma ,biology.protein ,medicine ,Cancer research ,Ex vivo ,Astrocyte - Abstract
The tumor microenvironment plays an essential role in supporting glioma stemness and radioresistance, and following radiotherapy, recurrent gliomas form in an irradiated microenvironment. Here, we found that astrocytes, when pre-irradiated, increased stemness and survival of co-cultured glioma cells. Tumor-naïve brains increased reactive astrocytes in response to radiation, and mice subjected to radiation prior to implantation of glioma cells developed more aggressive tumors. We identified extracellular matrix derived from irradiated astrocytes as a major driver of this phenotype, and astrocyte-derived transglutaminase 2 (TGM2) as a promoter of glioma stemness and radioresistance. TGM2 levels were increased after radiation in vivo and in recurrent human glioma, and TGM2 inhibitors abrogated glioma stemness and survival. These data suggest that irradiation of the brain results in the formation of a tumor-supportive microenvironment. Therapeutic targeting of radiation-induced, astrocyte-derived extracellular matrix proteins may enhance the efficacy of standard of care radiotherapy by reducing stemness in glioma.
- Published
- 2020
- Full Text
- View/download PDF
39. Influenza virus infection causes global RNAPII termination defects
- Author
-
Bahareh Haddad, Alex Rialdi, Ernesto Guccione, Jessica Sook Yuin Ho, Kimaada Allette, Natasha Moshkina, Minji Byun, Jian Jin, Adolfo García-Sastre, Sweta Ravisankar, Nevan J. Krogan, Randy A. Albrecht, Dalila Pinto, Rab K. Prinjha, Terrence M. Tumpey, Vittorio Sebastiano, Nacho Mena, Judd F. Hultquist, Alexander Tarakhovsky, Massimo Squatrito, Vyacheslav Yurchenko, Nan Zhao, Diana Low, Juan Ayllon, Ivan Marazzi, Yixuan Ma, Melissa Smith, Romain Fenouil, David Jimenez-Morales, Harm van Bakel, Benjamin Greenbaum, Maria Teresa Sánchez-Aparicio, and Robert Sebra
- Subjects
Terminator Regions, Genetic ,0301 basic medicine ,Virulence ,biology ,Cell ,SUMO protein ,RNA polymerase II ,medicine.disease_cause ,Virology ,Virus ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Downregulation and upregulation ,Influenza A virus ,Structural Biology ,Influenza, Human ,medicine ,biology.protein ,Humans ,RNA Polymerase II ,Molecular Biology ,Gene - Abstract
Viral infection perturbs host cells and can be used to uncover regulatory mechanisms controlling cellular responses and susceptibility to infections. Using cell biological, biochemical, and genetic tools, we reveal that influenza A virus (IAV) infection induces global transcriptional defects at the 3′ ends of active host genes and RNA polymerase II (RNAPII) run-through into extragenic regions. Deregulated RNAPII leads to expression of aberrant RNAs (3′ extensions and host-gene fusions) that ultimately cause global transcriptional downregulation of physiological transcripts, an effect influencing antiviral response and virulence. This phenomenon occurs with multiple strains of IAV, is dependent on influenza NS1 protein, and can be modulated by SUMOylation of an intrinsically disordered region (IDR) of NS1 expressed by the 1918 pandemic IAV strain. Our data identify a strategy used by IAV to suppress host gene expression and indicate that polymorphisms in IDRs of viral proteins can affect the outcome of an infection. Influenza A virus (IAV) infection induces transcription termination defects in host cells, an effect modulated by SUMOylation of an intrinsically disordered region of the influenza NS1 protein expressed by the 1918 pandemic IAV strain.
- Published
- 2018
- Full Text
- View/download PDF
40. TAMI-05. THE IRRADIATED BRAIN MICROENVIRONMENT SUPPORTS GLIOMA STEMNESS AND SURVIVAL VIA ASTROCYTE-DERIVED TRANSGLUTAMINASE 2
- Author
-
Håkan Axelson, Carolina Marques, Kristoffer von Stedingk, Valeria Governa, Vasiliki Pantazopoulou, Alexander Pietras, David Lindgren, Fredrik J. Swartling, Elin J. Pietras, Elinn Johansson, Johan Bengzon, Massimo Squatrito, Tobias Bergström, Tracy J. Berg, and Mattias Belting
- Subjects
Cancer Research ,biology ,Chemistry ,Tissue transglutaminase ,Tumor Microenvironment/Angiogenesis/Metabolism/Invasion ,medicine.disease ,medicine.anatomical_structure ,Oncology ,Glioma ,medicine ,biology.protein ,Cancer research ,Neurology (clinical) ,Astrocyte - Abstract
The highest-grade gliomas invariably recur as incurable tumors following standard of care comprising surgery, radiotherapy, and chemotherapy. The majority of the recurrent tumors form within the area of the brain receiving high-dose irradiation during treatment of the primary tumor, indicating that the recurrent tumor forms in an irradiated microenvironment. The tumor microenvironment has been demonstrated to influence the therapeutic response and stemness characteristics of tumor cells, but the influence of radiation on the microenvironment and its subsequent consequences for tumor cells are incompletely understood. Here, we used genetically engineered glioma mouse models and human glioma samples to characterize the impact of standard of care radiotherapy on the brain tumor microenvironment. We found that tumor-associated astrocytes subjected to radiation in vitro could enhance tumor cell stemness and survival of co-cultured glioma cells. More aggressive gliomas formed in vivo when mouse brains were irradiated prior to tumor cell implantation, suggesting that the irradiated brain microenvironment supports tumor growth. We isolated the effect of irradiated astrocytes to extracellular matrix secreted by these cells, and specifically found that astrocyte-derived transglutaminase 2 (TGM2) is a stromal promoter of glioma stemness and radioresistance. TGM2 levels were increased after radiation in glioma mouse models. Recombinant TGM2 enhanced, and TGM2 inhibitors blocked, glioma cell stemness. In human GBM tissue, TGM2 levels were increased in recurrent vs. primary tumors. In summary, in addition to supporting TGM2 as a potential therapeutic target in glioma, our data indicate that radiotherapy results in a tumor-supportive microenvironment, the targeting of which may be necessary to overcome tumor cell therapeutic resistance and recurrence.
- Published
- 2020
- Full Text
- View/download PDF
41. MGMT genomic rearrangements contribute to chemotherapy resistance in gliomas
- Author
-
Nuria Vaquero-Siguero, Marcos Galán-Ganga, Quanhua Mu, Tao Jiang, Zheng Wang, Hanjie Liu, Paula Kroon, Sandra Rodriguez-Perales, Junfei Zhao, Massimo Squatrito, Hoon Kim, Do Hyun Nam, Zhaoshi Bao, Raul Rabadan, Roel G.W. Verhaak, Jiguang Wang, Jason K. Sa, Ying Zhang, Barbara Oldrini, Seve Ballesteros Foundation, Asociación Española Contra el Cáncer, Natural Science Foundation of China (NSFC)/Research Grants Council (RGC), Beijing Municipal Administration of Hospitals Clinical Medicine Development of Special Funding Support, and Asociacion Espanola Contra el Cancer (AECC)
- Subjects
0301 basic medicine ,Male ,Methyltransferase ,Cancer therapy ,General Physics and Astronomy ,02 engineering and technology ,Drug resistance ,DNA Adducts ,Mice ,Cancer genomics ,CRISPR ,PROMOTER METHYLATION ,BENEFIT ,RNA-Seq ,RNA-SEQ ,lcsh:Science ,Promoter Regions, Genetic ,DNA Modification Methylases ,Gene Rearrangement ,Multidisciplinary ,Brain Neoplasms ,TEMOZOLOMIDE ,Glioma ,Middle Aged ,021001 nanoscience & nanotechnology ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,DNA methylation ,Biomarker (medicine) ,Female ,0210 nano-technology ,medicine.drug ,Adult ,Adolescent ,Science ,GLIOBLASTOMA ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Young Adult ,In vivo ,Cell Line, Tumor ,DNA adduct ,medicine ,Temozolomide ,Animals ,Humans ,neoplasms ,Aged ,Whole Genome Sequencing ,business.industry ,Tumor Suppressor Proteins ,General Chemistry ,Gene rearrangement ,DNA Methylation ,medicine.disease ,Xenograft Model Antitumor Assays ,GENE ,Microvesicles ,digestive system diseases ,nervous system diseases ,CNS cancer ,030104 developmental biology ,DNA Repair Enzymes ,Drug Resistance, Neoplasm ,Cancer research ,lcsh:Q ,Neoplasm Recurrence, Local ,business - Abstract
Temozolomide (TMZ) is an oral alkylating agent used for the treatment of glioblastoma and is now becoming a chemotherapeutic option in patients diagnosed with high-risk low-grade gliomas. The O-6-methylguanine-DNA methyltransferase (MGMT) is responsible for the direct repair of the main TMZ-induced toxic DNA adduct, the O6-Methylguanine lesion. MGMT promoter hypermethylation is currently the only known biomarker for TMZ response in glioblastoma patients. Here we show that a subset of recurrent gliomas carries MGMT genomic rearrangements that lead to MGMT overexpression, independently from changes in its promoter methylation. By leveraging the CRISPR/Cas9 technology we generated some of these MGMT rearrangements in glioma cells and demonstrated that the MGMT genomic rearrangements contribute to TMZ resistance both in vitro and in vivo. Lastly, we showed that such fusions can be detected in tumor-derived exosomes and could potentially represent an early detection marker of tumor recurrence in a subset of patients treated with TMZ., Chemotherapy resistance in recurrent gliomas is a large hurdle for successful therapy. Here, the authors show that some recurrent gliomas harbour O-6-methylguanine-DNA methyltransferase (MGMT) genomic rearrangements, and in vitro and in vivo these contribute to temozolomide resistance.
- Published
- 2020
42. LIF regulates CXCL9 in tumor-associated macrophages and prevents CD8+ T cell tumor-infiltration impairing anti-PD1 therapy
- Author
-
Carolina Marques, Juan Sahuquillo, Elena Garralda, Ester Bonfill-Teixidor, Isabel Cuartas, Josep Tabernero, Estela Pineda, Irene Brana, Maria Vieito, Francisco Martínez-Ricarte, Francesc Graus, Laura Escudero, Carlota Rubio-Perez, Mónica Pascual-García, Raffaella Iurlaro, Massimo Squatrito, Joan Seoane, Ester Planas-Rigol, Alexandra Arias, Paolo Nuciforo, Ada Sala-Hojman, Carmen Espejo, Leire Pedrosa, Isabel Huber-Ruano, European Research Council, Asociación Española Contra el Cáncer, Instituto de Salud Carlos III, FERO Foundation, Fundación La Caixa, Fundación BBVA, Cellex Foundation, Institut Català de la Salut, [Pascual-García M, Bonfill-Teixidor E] Vall d'Hebron Institut d'Oncologia, Barcelona, Spain. Hospital Universitari Vall d'Hebron, Barcelona, Spain. CIBERONC, Madrid, Spain. [Planas-Rigol E, Rubio-Perez C, Iurlaro R, Arias A, Cuartas I, Sala-Hojman A, Escudero L, Huber-Ruano I, Nuciforo P, Braña I, Garralda E, Vieito M] Vall d'Hebron Institut d'Oncologia, Barcelona, Spain. Hospital Universitari Vall d'Hebron, Barcelona, Spain. [Martínez-Ricarte F, Espejo C, Sahuquillo J] Vall d’Hebron Institut de Recerca, Barcelona, Spain. Hospital Universitari Vall d'Hebron, Barcelona, Spain. Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain. [Tabernero J] Vall d'Hebron Institut d'Oncologia, Barcelona, Spain. Hospital Universitari Vall d'Hebron, Barcelona, Spain. CIBERONC, Madrid, Spain. Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain. [Seoane J] Vall d'Hebron Institut d'Oncologia, Barcelona, Spain. Hospital Universitari Vall d'Hebron, Barcelona, Spain. CIBERONC, Madrid, Spain. Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain. Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain., Hospital Universitari Vall d'Hebron, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Amino Acids, Peptides, and Proteins::Peptides::Intercellular Signaling Peptides and Proteins::Cytokines::Leukemia Inhibitory Factor [CHEMICALS AND DRUGS] ,General Physics and Astronomy ,02 engineering and technology ,Mice, SCID ,CD8-Positive T-Lymphocytes ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,Chemokine CXCL9 ,Leukemia Inhibitory Factor ,Medicaments antineoplàstics ,Epigenesis, Genetic ,Neoplasms ,Tumor Microenvironment ,Cytotoxic T cell ,Otros calificadores::Otros calificadores::/inmunología [Otros calificadores] ,lcsh:Science ,reproductive and urinary physiology ,Chemokine CCL2 ,Cancer ,Otros calificadores::Otros calificadores::/tratamiento farmacológico [Otros calificadores] ,Multidisciplinary ,Chemistry ,021001 nanoscience & nanotechnology ,Citocines - Immunologia ,3. Good health ,medicine.anatomical_structure ,embryonic structures ,CXCL9 ,Neoplasias [ENFERMEDADES] ,0210 nano-technology ,Infiltration (medical) ,Cancer microenvironment ,endocrine system ,Cells::Blood Cells::Leukocytes::Leukocytes, Mononuclear::Lymphocytes::T-Lymphocytes::CD8-Positive T-Lymphocytes [ANATOMY] ,Science ,T cell ,CCL2 ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,Other subheadings::Other subheadings::/immunology [Other subheadings] ,medicine ,Animals ,Humans ,células::células sanguíneas::leucocitos::leucocitos mononucleares::linfocitos::linfocitos T::linfocitos T CD8-positivos [ANATOMÍA] ,Cèl·lules T - Immunologia ,Macrophages ,Tumors - Tractament ,General Chemistry ,Immunotherapy ,medicine.disease ,Antibodies, Neutralizing ,Immune checkpoint ,Neoplasms [DISEASES] ,Mice, Inbred C57BL ,030104 developmental biology ,Cancer research ,lcsh:Q ,aminoácidos, péptidos y proteínas::péptidos::péptidos y proteínas de señalización intercelular::citocinas::factor inhibidor de la leucemia [COMPUESTOS QUÍMICOS Y DROGAS] ,Immunologic Memory ,CD8 ,Neoplasm Transplantation - Abstract
Cancer response to immunotherapy depends on the infiltration of CD8+ T cells and the presence of tumor-associated macrophages within tumors. Still, little is known about the determinants of these factors. We show that LIF assumes a crucial role in the regulation of CD8+ T cell tumor infiltration, while promoting the presence of protumoral tumor-associated macrophages. We observe that the blockade of LIF in tumors expressing high levels of LIF decreases CD206, CD163 and CCL2 and induces CXCL9 expression in tumor-associated macrophages. The blockade of LIF releases the epigenetic silencing of CXCL9 triggering CD8+ T cell tumor infiltration. The combination of LIF neutralizing antibodies with the inhibition of the PD1 immune checkpoint promotes tumor regression, immunological memory and an increase in overall survival., LIF is a pleiotropic cytokine that promotes an immunosuppressive microenvironment and has critical functions in embryonic development. Here, the authors show that LIF regulates CD8+ T cell tumor infiltration in cancer by repressing CXCL19 and promoting the presence of protumoral macrophages and thatLIF inhibition, via neutralizing antibodies, promotes T cell infiltration and synergizes with immune checkpoint inhbitors resulting in tumor regression and immunological memory.
- Published
- 2019
43. NF1 Regulates Mesenchymal Glioblastoma Plasticity and Aggressiveness Through the AP-1 Transcription Factor FOSL1
- Author
-
Maria Stella Carro, Gaetano Gargiulo, Carolina Marques, Barbara Oldrini, Oliver Schnell, Annalisa Izzo, Eva Kling, Thomas Unterkircher, Sven Nelander, Yuliia Dramaretska, Paula Kroon, Massimo Squatrito, Erwin F. Wagner, and Latifa Bakiri
- Subjects
Cancer Research ,Mesenchymal Glioblastoma ,Mesenchymal stem cell ,Regulator ,Master regulator ,Gene signature ,Plasticity ,Biology ,FOSL1 ,Neural stem cell ,nervous system diseases ,Cell biology ,AP-1 transcription factor ,Cancer research ,Stem cell ,Transcription factor - Abstract
Summary The molecular basis underlying Glioblastoma (GBM) heterogeneity and plasticity are not fully understood. Using transcriptomic data of patient-derived brain tumor stem cell lines (BTSCs), classified based on GBM-intrinsic signatures, we identify the AP-1 transcription factor FOSL1 as a master regulator of the mesenchymal (MES) subtype. We provide a mechanistic basis to the role of the Neurofibromatosis type 1 gene ( NF1 ), a negative regulator of the RAS/MAPK pathway, in GBM mesenchymal transformation through the modulation of FOSL1 expression. Depletion of FOSL1 in NF1-mutant human BTSCs and Kras-mutant mouse neural stem cells results in loss of the mesenchymal gene signature, reduction in stem cell properties and in vivo tumorigenic potential. Our data demonstrate that FOSL1 controls GBM plasticity and aggressiveness in response to NF1 alterations. Significance Glioblastoma (GBM) is a very heterogenous disease for which multiple transcriptional subtypes have been described. Among these subtypes, the Mesenchymal (MES) GBMs have the worst prognosis. Here we provide the first causal evidence linking Neurofibromatosis type 1 gene (NF1) signalling and the acquisition of a MES gene expression program through the regulation of the AP-1 transcription factor FOSL1 . Using patient expression datasets, combined with in vitro and in vivo gain- and loss-of function mouse models, we show that FOSL1 is an important modulator of GBM that is required and sufficient for the activation of a MES program. Our work sheds light on the mechanisms that control the tumorigenicity of the most aggressive adult brain tumor type.
- Published
- 2019
- Full Text
- View/download PDF
44. Correction: Glioblastoma and glioblastoma stem cells are dependent on functional MTH1
- Author
-
Alberto J. Schuhmacher, Daniel Hägerstrand, Lars Bräutigam, Christina Kalderén, Pegah Rouhi, Linda Pudelko, Helge Gad, Massimo Squatrito, Ulrika Warpman Berglund, Steven J. Edwards, Kumar Sanjiv, Thomas Helleday, and Andreas Höglund
- Subjects
0303 health sciences ,business.industry ,Correction ,Biology ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,Text mining ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,Stem cell ,business ,030304 developmental biology ,Glioblastoma - Abstract
Glioblastoma multiforme (GBM) is an aggressive form of brain cancer with poor prognosis. Cancer cells are characterized by a specific redox environment that adjusts metabolism to its specific needs and allows the tumor to grow and metastasize. As a consequence, cancer cells and especially GBM cells suffer from elevated oxidative pressure which requires antioxidant-defense and other sanitation enzymes to be upregulated. MTH1, which degrades oxidized nucleotides, is one of these defense enzymes and represents a promising cancer target. We found MTH1 expression levels elevated and correlated with GBM aggressiveness and discovered that siRNA knock-down or inhibition of MTH1 with small molecules efficiently reduced viability of patient-derived GBM cultures. The effect of MTH1 loss on GBM viability was likely mediated through incorporation of oxidized nucleotides and subsequent DNA damage. We revealed that MTH1 inhibition targets GBM independent of aggressiveness as well as potently kills putative GBM stem cells
- Published
- 2020
- Full Text
- View/download PDF
45. TMOD-20. TRKing DOWN NOVEL THERAPEUTIC TARGETS IN GLIOMAS
- Author
-
Barbara Oldrini and Massimo Squatrito
- Subjects
Cancer Research ,Abstracts ,Oncology ,Neurology (clinical) - Abstract
The occurrence and importance of gene fusions in glioma has been appreciated only recently, largely due to high-throughput technologies, and gene fusions have been indicated as one of the major genomic abnormalities in glioblastoma (GBM), the most frequent and aggressive glioma subtype. The functional role of the majority of these alterations is completely unexplored. Recurrent gene fusions involving the Tropomyosin Receptor tyrosine Kinases (TRK) receptor family have been recently described in a variety of tumors, including both pediatric and adult low-grade (LGG) and high-grade gliomas (HGG). Strikingly, 40% of non-brain stem pediatric HGGs in infants have been shown to carry TRK gene fusions, making Trk inhibition an important potential therapeutic intervention in patients in which the current treatment modalities have devastating side effects. For the in vivo study of genomic rearrangements, we have recently generated an innovative mouse model that combines the genome editing capability of the CRISPR/Cas9 system with the somatic gene delivery of the RCAS/tv-a system. By searching the scientific literature we have identified approximately 30 different fusion involving NTRK1, NTRK2 and NTRK3 genes. To discriminate those fusions with tumorigenic potential in gliomas we have performed an in vivo gRNA pair screening. Four different RCAS-gRNA library pools have been transduced into p53-null TVA-Cas9 neural stem cells (NSCs) and subsequently transplanted into NOD/SCID mice. So far, half of the mice rapidly developed (1–2 months) quite aggressive tumors. We are currently characterizing those tumors to identify the most potent oncogenic NTRK fusions. NTRK gene rearrangements are emerging as novel targets across multiple tumor types, because of the increasing availability of new drugs with anti-Trk activity. As part of our studies, we are investigating the response and the resistance mechanism to Trk inhibitors that are currently used in different clinical trials.
- Published
- 2018
46. GlioVis data portal for visualization and analysis of brain tumor expression datasets
- Author
-
Angel Pazos Carro, Massimo Squatrito, Qianghu Wang, Robert L. Bowman, and Roel G.W. Verhaak
- Subjects
0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,Biomedical Research ,Statistics as Topic ,MEDLINE ,Brain tumor ,Computational biology ,Biology ,03 medical and health sciences ,Text mining ,Databases, Genetic ,medicine ,Humans ,Letter to the Editor ,Internet ,Brain Neoplasms ,business.industry ,medicine.disease ,Visualization ,Data portal ,030104 developmental biology ,Oncology ,Expression (architecture) ,Neurology (clinical) ,business - Published
- 2016
- Full Text
- View/download PDF
47. EGFR feedback-inhibition by Ran-binding protein 6 is disrupted in cancer
- Author
-
Ingo K. Mellinghoff, Craig M. Bielski, Maryam Pourmaleki, Paolo Codega, Maria Stella Carro, Massimo Squatrito, Barry S. Taylor, Álvaro Curiel-García, Paul Tempst, Daniel Rohle, John Blenis, Travis J. Hollmann, Barbara Oldrini, Igor Vivanco, Carl Campos, Hediye Erdjument-Bromage, Wan-Ying Hsieh, Marc K. Rosenblum, Christian Grommes, National Institutes of Health (Estados Unidos), Geoffrey Beene Foundation, Fundación Seve Ballesteros, and Memorial Sloan Kettering Cancer Center
- Subjects
STAT3 Transcription Factor ,0301 basic medicine ,Science ,Active Transport, Cell Nucleus ,General Physics and Astronomy ,Mice, SCID ,Importin ,medicine.disease_cause ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Gene silencing ,Epidermal growth factor receptor ,lcsh:Science ,STAT3 ,Cells, Cultured ,Feedback, Physiological ,Mice, Knockout ,Antibiotics, Antineoplastic ,Multidisciplinary ,biology ,Chemistry ,Glioma ,General Chemistry ,beta Karyopherins ,Xenograft Model Antitumor Assays ,3. Good health ,Cell biology ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,HEK293 Cells ,ran GTP-Binding Protein ,030104 developmental biology ,Doxorubicin ,Gene Knockdown Techniques ,biology.protein ,STAT protein ,lcsh:Q ,Female ,Beta Karyopherins ,Nuclear transport ,Carcinogenesis - Abstract
Transport of macromolecules through the nuclear pore by importins and exportins plays a critical role in the spatial regulation of protein activity. How cancer cells co-opt this process to promote tumorigenesis remains unclear. The epidermal growth factor receptor (EGFR) plays a critical role in normal development and in human cancer. Here we describe a mechanism of EGFR regulation through the importin β family member RAN-binding protein 6 (RanBP6), a protein of hitherto unknown functions. We show that RanBP6 silencing impairs nuclear translocation of signal transducer and activator of transcription 3 (STAT3), reduces STAT3 binding to the EGFR promoter, results in transcriptional derepression of EGFR, and increased EGFR pathway output. Focal deletions of the RanBP6 locus on chromosome 9p were found in a subset of glioblastoma (GBM) and silencing of RanBP6 promoted glioma growth in vivo. Our results provide an example of EGFR deregulation in cancer through silencing of components of the nuclear import pathway., The epidermal growth factor receptor (EGFR) signalling is regulated at multiple levels. Here the authors show that the importin RanBP6 acts as a tumor suppressor in Glioblastoma and regulates EGFR signalling through promoting translocation of STAT3 to the nuclei and repressing EGFR transcription.
- Published
- 2017
- Full Text
- View/download PDF
48. TMIC-22. DECIPHERING GLIOMA INTRINSIC TRANSCRIPTIONAL SUBTYPES IDENTIFIES TUMOR EVOLUTION ASSOCIATES WITH CHANGES IN IMMUNE-MICROENVIRONMENT
- Author
-
Jian Hu, Joanna J. Phillips, Qianghu Wang, Floris P. Barthel, Amy B. Heimberger, Yan Li, Siyuan Zheng, Konrad Gabrusiewicz, Yu-Hsi Lin, Baoli Hu, Nikuj Satani, Erik P. Sulman, Pengping Li, Florian L. Muller, Ana C. deCarvalho, Guocan Wang, Gaetano Finocchiaro, Xin Hu, Sali Lyu, Massimo Squatrito, Edward F. Chang, Adriana Olar, Do-Hyun Nam, Zhengdao Lan, Tom Mikkelsen, Emmanuel Martinez-Ledesma, Roel G.W. Verhaak, Ronald A. DePinho, Eskil Eskilsson, Mitchel S. Berger, Hee Jin Cho, and Lisa Scarpace
- Subjects
0301 basic medicine ,03 medical and health sciences ,Cancer Research ,Abstracts ,030104 developmental biology ,Oncology ,Glioma ,Immune microenvironment ,Cancer research ,medicine ,Neurology (clinical) ,Biology ,medicine.disease - Abstract
Glioblastoma expression subtypes have been previously been associated with genomic abnormalities, treatment response, and differences in tumor microenvironment. We leveraged IDH wild-type glioblastomas, derivative neurospheres, and single cell gene expression profiles to define three tumor-intrinsic transcriptional subtypes designated as proneural, mesenchymal, and classical, a revision of the previously reported TCGA subtypes. Transcriptomic subtype multiplicity correlated with increased intratumoral heterogeneity and the presence of tumor microenvironment. In silico cell sorting identified macrophages/microglia, CD4+ T lymphocytes, and neutrophils in the glioma microenvironment. NF1 deficiency resulted in increased tumor-associated macrophages/microglia infiltration. Comparison of matching primary and recurrent gliomas elucidated treatment-induced phenotypic tumor evolution, including expression subtype switching, in 45% of our cohort as well as associations between microenvironmental components and treatment response. Gene signature-based tumor microenvironment inference revealed a decrease in invading monocytes and a subtype-dependent increase in macrophages/microglia cells upon disease recurrence. Hypermutation at diagnosis or at recurrence was associated with CD8+ T cell enrichment. Frequency of M2 macrophage detection was associated with short-term relapse after radiation therapy. Our study provides a comprehensive transcriptional and cellular landscape of IDH wild-type glioblastoma during treatment modulated tumor evolution. Characterization of the evolving glioblastoma transcriptome and tumor microenvironment aids in designing more effective immunotherapy trials.
- Published
- 2017
49. Somatic genome editing with the RCAS/TVA-CRISPR/Cas9 system for precision tumor modeling
- Author
-
Álvaro Curiel-García, Carolina Marques, Veronica Matia, Barbara Oldrini, Sandra Rodriguez-Perales, Özge Uluçkan, Massimo Squatrito, Jason T. Huse, and Raúl Torres-Ruiz
- Subjects
Homology directed repair ,Genetics ,Mutation ,Genome editing ,Cas9 ,CDKN2A ,Somatic cell ,medicine ,CRISPR ,Biology ,medicine.disease_cause ,Chromosomal Deletion - Abstract
It has been gradually established that the vast majority of human tumors are extraordinarily heterogeneous at a genetic level. To accurately recapitulate this complexity, it is now evident that in vivo animal models of cancers will require to recreate not just a handful of simple genetic alterations, but possibly dozens and increasingly intricate. Here, we have combined the RCAS/TVA system with the CRISPR/Cas9 genome editing tools for precise modeling of human tumors. We show that somatic deletion in neural stem cells (NSCs) of a variety of known tumor suppressor genes (Trp53, Cdkn2a and Pten), in combination with the expression of an oncogene driver, leads to high-grade glioma formation. Moreover, by simultaneous delivery of pairs of guide RNAs (gRNAs) we generated different gene fusions, either by chromosomal deletion (Bcan-Ntrk1) or by chromosomal translocation (Myb-Qk), and we show that they have transforming potential in vitro and in vivo. Lastly, using homology-directed-repair (HDR), we also produced tumors carrying the Braf V600E mutation, frequently identified in a variety of subtypes of gliomas. In summary, we have developed an extremely powerful and versatile mouse model for in vivo somatic genome editing, that will elicit the generation of more accurate cancer models particularly appropriate for pre-clinical testing.
- Published
- 2017
- Full Text
- View/download PDF
50. Tumor Evolution of Glioma-Intrinsic Gene Expression Subtypes Associates with Immunological Changes in the Microenvironment
- Author
-
Baoli Hu, Massimo Squatrito, Nikunj Satani, Eskil Eskilsson, Jian Hu, Charles Etienne Gabriel Sauvé, Mitchel S. Berger, Emmanuel Martinez-Ledesma, Roel G.W. Verhaak, Hoon Kim, Amy B. Heimberger, Yan Li, Siyuan Zheng, Konrad Gabrusiewicz, Do Hyun Nam, Joanna J. Phillips, Ronald A. DePinho, Hee Jin Cho, Floris P. Barthel, Edward I. Chang, Gaetano Finocchiaro, Florian L. Muller, Zheng D. Lan, Adriana Olar, Qianghu Wang, Ana C. deCarvalho, Pengping Li, Erik P. Sulman, Tom Mikkelsen, Guocan Wang, Sali Lyu, Yu Hsi Lin, Lisa Scarpace, Xin Hu, and Neurosurgery
- Subjects
0301 basic medicine ,Cancer Research ,tumor evolution ,T-Lymphocytes ,0302 clinical medicine ,Recurrence ,Gene expression ,Tumor Microenvironment ,2.1 Biological and endogenous factors ,Aetiology ,Cancer ,Regulation of gene expression ,Microglia ,mesenchymal subtype ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Phenotype ,Oncology ,030220 oncology & carcinogenesis ,T cell ,Oncology and Carcinogenesis ,Biology ,Article ,03 medical and health sciences ,disease recurrence ,immune cells ,Rare Diseases ,Glioma ,medicine ,Genetics ,Humans ,Oncology & Carcinogenesis ,macrophages/microglia ,Tumor microenvironment ,Neoplastic ,Gene Expression Profiling ,glioblastoma ,Neurosciences ,Cell Biology ,Gene signature ,medicine.disease ,Survival Analysis ,Brain Disorders ,Gene expression profiling ,proneural to mesenchymal transition ,Brain Cancer ,030104 developmental biology ,Gene Expression Regulation ,Cancer cell ,Cancer research ,Glioblastoma ,CD8 - Abstract
We leveraged IDH wild-type glioblastomas, derivative neurospheres, and single-cell gene expression profiles to define three tumor-intrinsic transcriptional subtypes designated as proneural, mesenchymal, and classical. Transcriptomic subtype multiplicity correlated with increased intratumoral heterogeneity and presence of tumor microenvironment. In silico cell sorting identified macrophages/microglia, CD4+ T lymphocytes, and neutrophils in the glioma microenvironment. NF1 deficiency resulted in increased tumor-associated macrophages/microglia infiltration. Longitudinal transcriptome analysis showed that expression subtype is retained in 55% of cases. Gene signature-based tumor microenvironment inference revealed a decrease in invading monocytes and a subtype-dependent increase in macrophages/microglia cells upon disease recurrence. Hypermutation at diagnosis or at recurrence associated with CD8+ Tcell enrichment. Frequency of M2 macrophages detection associated with short-term relapse after radiation therapy.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.