Your browser doesn't support javascript.
loading
Selective inhibition of protein arginine methyltransferase 5 blocks initiation and maintenance of B-cell transformation.
Alinari, Lapo; Mahasenan, Kiran V; Yan, Fengting; Karkhanis, Vrajesh; Chung, Ji-Hyun; Smith, Emily M; Quinion, Carl; Smith, Porsha L; Kim, Lisa; Patton, John T; Lapalombella, Rosa; Yu, Bo; Wu, Yun; Roy, Satavisha; De Leo, Alessandra; Pileri, Stefano; Agostinelli, Claudio; Ayers, Leona; Bradner, James E; Chen-Kiang, Selina; Elemento, Olivier; Motiwala, Tasneem; Majumder, Sarmila; Byrd, John C; Jacob, Samson; Sif, Said; Li, Chenglong; Baiocchi, Robert A.
Affiliation
  • Alinari L; Division of Hematology, Department of Internal Medicine.
  • Mahasenan KV; Division of Medicinal Chemistry and Pharmacognosy, and.
  • Yan F; Division of Hematology, Department of Internal Medicine.
  • Karkhanis V; Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH;
  • Chung JH; Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH;
  • Smith EM; Division of Hematology, Department of Internal Medicine.
  • Quinion C; Division of Hematology, Department of Internal Medicine.
  • Smith PL; Division of Hematology, Department of Internal Medicine.
  • Kim L; Division of Hematology, Department of Internal Medicine.
  • Patton JT; Division of Hematology, Department of Internal Medicine.
  • Lapalombella R; Division of Hematology, Department of Internal Medicine.
  • Yu B; Division of Hematology, Department of Internal Medicine.
  • Wu Y; Division of Hematology, Department of Internal Medicine.
  • Roy S; Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH;
  • De Leo A; Department of Public Health and Infectious Diseases, University La Sapienza, Rome, Italy;
  • Pileri S; Hematopathology Unit, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy;
  • Agostinelli C; Hematopathology Unit, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy;
  • Ayers L; Department of Pathology, The Ohio State University, Columbus, OH;
  • Bradner JE; Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA;
  • Chen-Kiang S; Department of Pathology and Laboratory Medicine, and.
  • Elemento O; Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY; and.
  • Motiwala T; Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH;
  • Majumder S; Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH;
  • Byrd JC; Division of Hematology, Department of Internal Medicine, Division of Medicinal Chemistry and Pharmacognosy, and.
  • Jacob S; Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH;
  • Sif S; Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar.
  • Li C; Division of Medicinal Chemistry and Pharmacognosy, and.
  • Baiocchi RA; Division of Hematology, Department of Internal Medicine.
Blood ; 125(16): 2530-43, 2015 Apr 16.
Article in En | MEDLINE | ID: mdl-25742700
ABSTRACT
Epigenetic events that are essential drivers of lymphocyte transformation remain incompletely characterized. We used models of Epstein-Barr virus (EBV)-induced B-cell transformation to document the relevance of protein arginine methyltransferase 5 (PRMT5) to regulation of epigenetic-repressive marks during lymphomagenesis. EBV(+) lymphomas and transformed cell lines exhibited abundant expression of PRMT5, a type II PRMT enzyme that promotes transcriptional silencing of target genes by methylating arginine residues on histone tails. PRMT5 expression was limited to EBV-transformed cells, not resting or activated B lymphocytes, validating it as an ideal therapeutic target. We developed a first-in-class, small-molecule PRMT5 inhibitor that blocked EBV-driven B-lymphocyte transformation and survival while leaving normal B cells unaffected. Inhibition of PRMT5 led to lost recruitment of a PRMT5/p65/HDAC3-repressive complex on the miR96 promoter, restored miR96 expression, and PRMT5 downregulation. RNA-sequencing and chromatin immunoprecipitation experiments identified several tumor suppressor genes, including the protein tyrosine phosphatase gene PTPROt, which became silenced during EBV-driven B-cell transformation. Enhanced PTPROt expression following PRMT5 inhibition led to dephosphorylation of kinases that regulate B-cell receptor signaling. We conclude that PRMT5 is critical to EBV-driven B-cell transformation and maintenance of the malignant phenotype, and that PRMT5 inhibition shows promise as a novel therapeutic approach for B-cell lymphomas.
Subject(s)

Full text: 1 Database: MEDLINE Main subject: Protein-Arginine N-Methyltransferases / B-Lymphocytes / Cell Transformation, Viral / Enzyme Inhibitors Type of study: Prognostic_studies Language: En Year: 2015 Type: Article

Full text: 1 Database: MEDLINE Main subject: Protein-Arginine N-Methyltransferases / B-Lymphocytes / Cell Transformation, Viral / Enzyme Inhibitors Type of study: Prognostic_studies Language: En Year: 2015 Type: Article