Loss of the BCR-FGFR1 GEF Domain Suppresses RHOA Activation and Enhances B-Lymphomagenesis in Mice.

Transformation of hematopoietic stem cells by the BCR-FGFR1 fusion kinase found in a variant of stem cell leukemia/lymphoma (SCLL) syndrome leads to development of B-lymphomas in syngeneic mice and humans. In this study, we show that the relatively rapid onset of this leukemia is potentially related to oncogenic domains within the BCR component. BCR recruited a guanidine nucleotide exchange factor (GEF) domain to the fusion kinase to facilitate activation of small GTPases such as the Ras homology gene family, member A (RHOA). Deletion of this GEF domain increased leukemogenesis, enhanced cell survival and proliferation, and promoted stem cell expansion and lymph node metastasis. This suggests that, in an SCLL context, the presence of the endogenous GEF motif leads to reduced leukemogenesis. Indeed, loss of the GEF domain suppressed activation of RHOA and PTEN, leading to increased activation of AKT. Loss of the GEF domain enhanced cell proliferation and invasion potential, which was also observed in cells in which RHOA is knocked down, supported by the observation that overexpression of RHOA leads to reduced viability and invasion. In vivo depletion of RHOA in SCLL cells significantly increased disease progression and shortened latency. Collectively, these data show that the BCR GEF domain affects phenotypes associated with progression of SCLL through suppression of RHOA signaling. SIGNIFICANCE: RHOA activation is a critical event in the progression of BCR-FGFR1-driven leukemogenesis in stem cell leukemia and lymphoma syndrome and is regulated by the BCR GEF domain.

[1]  E. Kitamura,et al.  The miR-17/92 cluster is involved in the molecular etiology of the SCLL syndrome driven by the BCR-FGFR1 chimeric kinase , 2017, Oncogene.

[2]  L. Medeiros,et al.  Myeloproliferative neoplasms with t(8;22)(p11.2;q11.2)/BCR-FGFR1: a meta-analysis of 20 cases shows cytogenetic progression with B-lymphoid blast phase. , 2017, Human pathology.

[3]  A. Pandey,et al.  Differential signaling through p190 and p210 BCR-ABL fusion proteins revealed by interactome and phosphoproteome analysis , 2017, Leukemia.

[4]  O. Hantschel,et al.  Differential signaling networks of Bcr–Abl p210 and p190 kinases in leukemia cells defined by functional proteomics , 2017, Leukemia.

[5]  E. Kitamura,et al.  A model of BCR‐FGFR1 driven human AML in immunocompromised mice , 2016, British journal of haematology.

[6]  Edd Ricker,et al.  Faculty of 1000 evaluation for Rho-associated coiled-coil containing kinases (ROCK): structure, regulation, and functions. , 2016 .

[7]  S. Malek,et al.  Targeting FGFR1 to suppress leukemogenesis in syndromic and de novo AML in murine models , 2016, Oncotarget.

[8]  J. Zehnder,et al.  Clinical activity of ponatinib in a patient with FGFR1-rearranged mixed-phenotype acute leukemia , 2016, Leukemia.

[9]  O. Nureki,et al.  Variegated RHOA mutations in adult T-cell leukemia/lymphoma. , 2016, Blood.

[10]  N. Itoh,et al.  The Fibroblast Growth Factor signaling pathway , 2015, Wiley interdisciplinary reviews. Developmental biology.

[11]  M. Olson,et al.  Rho-associated coiled-coil containing kinases (ROCK) , 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]  S. Narumiya,et al.  Physiological roles of Rho and Rho effectors in mammals. , 2013, European journal of cell biology.

[16]  K. Green,et al.  The GEF Bcr activates RhoA/MAL signaling to promote keratinocyte differentiation via desmoglein-1 , 2013, The Journal of cell biology.

[17]  David A. Williams,et al.  Contributions of the RhoGEF activity of p210 BCR/ABL to disease progression , 2012, Leukemia.

[18]  J. Cowell,et al.  Ponatinib suppresses the development of myeloid and lymphoid malignancies associated with FGFR1 abnormalities , 2012, Leukemia.

[19]  J. Cowell,et al.  Acute Progression of BCR-FGFR1 Induced Murine B-Lympho/Myeloproliferative Disorder Suggests Involvement of Lineages at the Pro-B Cell Stage , 2012, PloS one.

[20]  J. Cowell,et al.  Constitutive Notch pathway activation in murine ZMYM2-FGFR1-induced T-cell lymphomas associated with atypical myeloproliferative disease. , 2011, Blood.

[21]  K. Kaibuchi,et al.  Rho-Kinase/ROCK: A Key Regulator of the Cytoskeleton and Cell Polarity , 2010, Cytoskeleton.

[22]  L. Medeiros,et al.  8p11 myeloproliferative syndrome: a review. , 2010, Human pathology.

[23]  N. Turner,et al.  Fibroblast growth factor signalling: from development to cancer , 2010, Nature Reviews Cancer.

[24]  Xiurong Li,et al.  Genetic fingerprinting of the development and progression of T-cell lymphoma in a murine model of atypical myeloproliferative disorder initiated by the ZNF198-fibroblast growth factor receptor-1 chimeric tyrosine kinase. , 2009, Blood.

[25]  P. L. Rodriguez,et al.  The RhoGEF domain of p210 Bcr-Abl activates RhoA and is required for transformation , 2008, Oncogene.

[26]  David A. Williams,et al.  Inhibition of RhoA GTPase activity enhances hematopoietic stem and progenitor cell proliferation and engraftment. , 2006, Blood.

[27]  Alan Hall,et al.  Rho GTPases: biochemistry and biology. , 2005, Annual review of cell and developmental biology.

[28]  Andrei V Bakin,et al.  WAVE3 promotes cell motility and invasion through the regulation of MMP-1, MMP-3, and MMP-9 expression. , 2005, Experimental cell research.

[29]  D. Birnbaum,et al.  Dual lympho-myeloproliferative disorder in a patient with t(8;22) with BCR-FGFR1 gene fusion. , 2005, International journal of oncology.

[30]  Rong Zeng,et al.  Regulation of PTEN by Rho small GTPases , 2005, Nature Cell Biology.

[31]  R. V. van Etten,et al.  Distinct stem cell myeloproliferative/T lymphoma syndromes induced by ZNF198-FGFR1 and BCR-FGFR1 fusion genes from 8p11 translocations. , 2004, Cancer cell.

[32]  J. Cowell,et al.  The Oncogenic Fusion Protein-tyrosine Kinase ZNF198/Fibroblast Growth Factor Receptor-1 Has Signaling Function Comparable with Interleukin-6 Cytokine Receptors* , 2003, The Journal of Biological Chemistry.

[33]  J. Melo,et al.  The t(8;22) in chronic myeloid leukemia fuses BCR to FGFR1: transforming activity and specific inhibition of FGFR1 fusion proteins. , 2001, Blood.