Activating mutations and translocations in the guanine exchange factor VAV1 in peripheral T-cell lymphomas

Significance Guanine nucleotide exchange factor VAV1 encodes an adaptor and signal transduction factor with important roles in T-cell receptor signaling. This study identifies activating VAV1 recurrent mutations and VAV1 fusions in peripheral T-cell lymphomas, directly establishing an oncogenic role for constitutive VAV1 signaling in the pathogenesis of this disease. Peripheral T-cell lymphomas (PTCLs) are a heterogeneous group of non-Hodgkin lymphomas frequently associated with poor prognosis and for which genetic mechanisms of transformation remain incompletely understood. Using RNA sequencing and targeted sequencing, here we identify a recurrent in-frame deletion (VAV1 Δ778–786) generated by a focal deletion-driven alternative splicing mechanism as well as novel VAV1 gene fusions (VAV1-THAP4, VAV1-MYO1F, and VAV1-S100A7) in PTCL. Mechanistically these genetic lesions result in increased activation of VAV1 catalytic-dependent (MAPK, JNK) and non–catalytic-dependent (nuclear factor of activated T cells, NFAT) VAV1 effector pathways. These results support a driver oncogenic role for VAV1 signaling in the pathogenesis of PTCL.

[1]  Sarah H. Johnson,et al.  Integrated mate-pair and RNA sequencing identifies novel, targetable gene fusions in peripheral T-cell lymphoma. , 2016, Blood.

[2]  Sven Diederichs,et al.  The dark matter of the cancer genome: aberrations in regulatory elements, untranslated regions, splice sites, non‐coding RNA and synonymous mutations , 2016, EMBO molecular medicine.

[3]  Yali Xue,et al.  BCFtools/RoH: a hidden Markov model approach for detecting autozygosity from next-generation sequencing data , 2016, Bioinform..

[4]  H. Aburatani,et al.  Integrated molecular analysis of adult T cell leukemia/lymphoma , 2015, Nature Genetics.

[5]  S. Katzav Vav1: A Dr. Jekyll and Mr. Hyde protein – good for the hematopoietic system, bad for cancer , 2015, Oncotarget.

[6]  A. Valencia,et al.  Non-coding recurrent mutations in chronic lymphocytic leukaemia , 2015, Nature.

[7]  J. Armitage The aggressive peripheral T‐cell lymphomas: 2015 , 2015, American journal of hematology.

[8]  A. Rosenwald,et al.  Convergent mutations and kinase fusions lead to oncogenic STAT3 activation in anaplastic large cell lymphoma. , 2015, Cancer cell.

[9]  Chris Wiggins,et al.  Pegasus: a comprehensive annotation and prediction tool for detection of driver gene fusions in cancer , 2014, BMC Systems Biology.

[10]  G. Montoya,et al.  The C-Terminal SH3 Domain Contributes to the Intramolecular Inhibition of Vav Family Proteins , 2014, Science Signaling.

[11]  X. Bustelo Vav family exchange factors: an integrated regulatory and functional view , 2014, Small GTPases.

[12]  Min Kyung Sung,et al.  A recurrent inactivating mutation in RHOA GTPase in angioimmunoblastic T cell lymphoma , 2014, Nature Genetics.

[13]  O. Nureki,et al.  Somatic RHOA mutation in angioimmunoblastic T cell lymphoma , 2014, Nature Genetics.

[14]  I. Lossos,et al.  Recurrent mutations in epigenetic regulators, RHOA and FYN kinase in peripheral T cell lymphomas , 2014, Nature Genetics.

[15]  Raul Rabadan,et al.  SAVI: a statistical algorithm for variant frequency identification , 2013, BMC Systems Biology.

[16]  B. Nathwani,et al.  Clinicopathologic characteristics of angioimmunoblastic T-cell lymphoma: analysis of the international peripheral T-cell lymphoma project. , 2013, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[17]  J. Armitage,et al.  The aggressive peripheral T‐cell lymphomas: 2012 update on diagnosis, risk stratification, and management , 2012, American journal of hematology.

[18]  M. Oberley,et al.  Vav1 in hematologic neoplasms, a mini review. , 2012, American journal of blood research.

[19]  Christopher A. Maher,et al.  ChimeraScan: a tool for identifying chimeric transcription in sequencing data , 2011, Bioinform..

[20]  K. Savage,et al.  Peripheral T-cell lymphoma, not otherwise specified. , 2017, Blood.

[21]  P. Gaulard,et al.  Pathology and biology of peripheral T‐cell lymphomas , 2011, Histopathology.

[22]  M. DePristo,et al.  The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. , 2010, Genome research.

[23]  John S. Olson,et al.  The structure and NO binding properties of the nitrophorin‐like heme‐binding protein from Arabidopsis thaliana gene locus At1g79260.1 , 2010, Proteins.

[24]  Michael K. Rosen,et al.  Structural and Energetic Mechanisms of Cooperative Autoinhibition and Activation of Vav1 , 2010, Cell.

[25]  S. Smerdon,et al.  Function of the Nucleotide Exchange Activity of Vav1 in T Cell Development and Activation , 2009, Science Signaling.

[26]  X. Bustelo,et al.  The Use of Knockout Mice Reveals a Synergistic Role of the Vav1 and Rasgrf2 Gene Deficiencies in Lymphomagenesis and Metastasis , 2009, PloS one.

[27]  D. Kioussis,et al.  Rac GTPases play critical roles in early T-cell development. , 2009, Blood.

[28]  Cui-zhu Zhang,et al.  The Calponin Homology Domain of Vav1 Associates with Calmodulin and Is Prerequisite to T Cell Antigen Receptor-induced Calcium Release in Jurkat T Lymphocytes* , 2007, Journal of Biological Chemistry.

[29]  Kathleen C. Lee,et al.  S100A7 (Psoriasin): a story of mice and men. , 2006, The Journal of investigative dermatology.

[30]  Somasekar Seshagiri,et al.  Somatic mutations lead to an oncogenic deletion of met in lung cancer. , 2006, Cancer research.

[31]  V. Tybulewicz,et al.  Vav-family proteins in T-cell signalling. , 2005, Current opinion in immunology.

[32]  J. Chernoff,et al.  Vav1 Transduces T Cell Receptor Signals to the Activation of the Ras/ERK Pathway via LAT, Sos, and RasGRP1* , 2004, Journal of Biological Chemistry.

[33]  H. Müller-Hermelink,et al.  Die WHO-Klassifikation maligner Lymphome , 2002, Der Radiologe.

[34]  X. Bustelo,et al.  Structural Determinants for the Biological Activity of Vav Proteins* , 2002, The Journal of Biological Chemistry.

[35]  A. Duncan,et al.  Pleiotropic defects in TCR signaling in a Vav‐1‐null Jurkat T‐cell line , 2002, The EMBO journal.

[36]  D. Kioussis,et al.  Vav1 Transduces T Cell Receptor Signals to the Activation of Phospholipase C-γ1 via Phosphoinositide 3-Kinase-dependent and -independent Pathways , 2002, The Journal of experimental medicine.

[37]  L. Chasin,et al.  Human Genomic Sequences That Inhibit Splicing , 2000, Molecular and Cellular Biology.

[38]  A. Weiss,et al.  A Guanine Nucleotide Exchange Factor-independent Function of Vav1 in Transcriptional Activation* , 2000, The Journal of Biological Chemistry.

[39]  M. Turner,et al.  The Rho-family GTP exchange factor Vav is a critical transducer of T cell receptor signals to the calcium, ERK, and NF-kappaB pathways. , 1999, Proceedings of the National Academy of Sciences of the United States of America.

[40]  J. Penninger,et al.  Vav Regulates Peptide-specific Apoptosis in Thymocytes , 1998, The Journal of experimental medicine.

[41]  F. Alt,et al.  Defects in actin-cap formation in Vav-deficient mice implicate an actin requirement for lymphocyte signal transduction , 1998, Current Biology.

[42]  K. Tedford,et al.  Vav is a regulator of cytoskeletal reorganization mediated by the T-cell receptor , 1998, Current Biology.

[43]  R. Zamoyska,et al.  A requirement for the Rho-family GTP exchange factor Vav in positive and negative selection of thymocytes. , 1997, Immunity.

[44]  C. Fizames,et al.  Cloning of the genes encoding two murine and human cochlear unconventional type I myosins. , 1997, Genomics.

[45]  K. Schuebel,et al.  Phosphotyrosine-dependent activation of Rac-1 GDP/GTP exchange by the vav proto-oncogene product , 1997, Nature.

[46]  J. Wu,et al.  A functional T-cell receptor signaling pathway is required for p95vav activity , 1995, Molecular and cellular biology.

[47]  F. Alt,et al.  Defective signalling through the T- and B-cell antigen receptors in lymphoid cells lacking the vav proto-oncogene , 1995, Nature.

[48]  K. Rajewsky,et al.  Defective antigen receptor-mediated proliferation of B and T cells in the absence of Vav , 1995, Nature.

[49]  J. Cleveland,et al.  Loss of the amino-terminal helix-loop-helix domain of the vav proto-oncogene activates its transforming potential , 1991, Molecular and cellular biology.

[50]  M. Barbacid,et al.  vav, a novel human oncogene derived from a locus ubiquitously expressed in hematopoietic cells. , 1989, The EMBO journal.

[51]  A. J. Lukes Classification of malignant lymphomas. , 1981, The Western journal of medicine.