Patrick L. Green

PhD

Robert H. Rainier Chair in Industrial Veterinary Medicine and Research
Professor and Associate Dean for Research and Graduate Studies
Associate Director for Basic Sciences at The Ohio State University Comprehensive Cancer Center
Director of the Center for Retrovirus Research

Contact

green.466@osu.edu (614) 292-9203 Veterinary Medicine Academic Building
1900 Coffey Road
Columbus, OH 43210
Map Link

Department

Veterinary Biosciences

Center For Retrovirus Research

Professional Training and experience

  • PhD (Oncology), University of Wisconsin - McArdle Laboratory for Cancer Research
  • Post-doctoral, University of California Los Angeles
  • Assistant Research Biologist, University of California, Los Angeles
  • Assistant Professor, Vanderbilt University
  • Associate Professor, The Ohio State University
  • Professor, The Ohio State University
  • Director, Center for Retrovirus Research, The Ohio State University
  • Associate Director, Basic Sciences, The Ohio State University Comprehensive Cancer Center

Research Interests

  • Molecular Pathogenesis of Human T-cell Leukemia Virus (HTLV)
  • Molecular Biology of Retrovirus Replication
  • T-cell Activation/Transformation


Research Projects

The laboratory of Patrick Green is internationally recognized for their contributions to the understanding of the molecular basis of T-lymphocyte transformation and induction of leukemia/lymphoma and neurological disease by the human T-cell leukemia viruses (HTLVs). The Green lab has three areas of research focus investigating viral and cellular regulators of HTLV gene expression/replication, cellular transformation, and virus survival or persistence in the infected host.
Focus/Project 1

One aim of our laboratory is to understand the mechanism of action and biological role of a unique HTLV-1 accessory/regulatory gene, termed Hbz. It is the only gene transcribed from the antisense strand of the viral proviral genome and is expressed in all adult T-cell leukemia (ATL) cells, whereas tax oncogene expression is typically undetectable. We have discovered that HBZ is dispensable in cell culture, but is required to enhance virus replication and survival in the infected host. More recently utilizing a Hbz shRNA knockdown approach and NOD/SCIDγc-/-(NOG) mice we discovered that Hbz expression enhances the proliferative capacity of HTLV-1 infected cells in culture and plays a critical role in infected cell survival and ultimately HTLV-1 tumorigenesis. HBZ, originally thought to be unique to HTLV-1 has been hypothesized to play a role in pathogenesis. Our overall hypothesis is that uncovering the mechanism of actions of Hbz and defining the interplay between Hbz and the viral transactivator, Tax, will provide important insight into HTLV-1 cellular transformation and disease and ultimately will provide means for therapeutic targeting to erradicate HTLV-1 persistence in the host.

We will dissect the mechanisms of action of Hbz focusing on Hbz mRNA activities, the functional role of HBZ post-translational modification, and effects on cellular protein interactions and pathways with emphasis on Jun, and NFκB and interferon (IRF-1, IRF-7) regulation. We will combine in vitro (transformation assays) and in vivo (transgenic and humanized mice) approaches to determine the interplay between Hbz and Tax in the cellular transformation and tumor induction process. Recently, an antisense protein of HTLV-2 (APH-2) was identified. Despite its lack of a typical b-ZIP domain, APH-2, like HBZ, interacts with CREB and down-regulates Tax-mediated viral transcription. We will investigate in detail the mechanism of action of APH-2. Importantly, since HTLV-1 and HTLV-2 are closely related retroviruses, but have distinct etiological roles in human disease, we hypothesize that comparative studies on anti-sense proteins of HTLV-1 and HTLV-2 will provide fundamental insights into their distinct pathogenic properties.
Focus/Project 2

Recently, our research team has focused to address the relevance of the arginine methyltransferase enzyme, PRMT5, to malignant disease and investigate the therapeutic potential of a new class of drugs to inhibit this enzyme. PRMT5 is an epigenetic modifier that silences the transcription of many key regulatory cellular genes through symmetric di-methylation (S2Me) of arginine (R) residues on histone proteins (H4R3 & H3R8). We are testing the hypothesis that PRMT5 contributes to HTLV-1-driven cellular transformation and leukemia, and ultimately determine the efficacy of PRMT5 inhibitors (PRMT5i) in a preclinical ATL mouse model. We found that PRMT5 expression was upregulated during HTLV-1-mediated T-cell transformation, as well as in established lymphocytic leukemia/lymphoma cell lines and ATLL patient PBMCs. shRNA-mediated reduction in PRMT5 protein levels or its inhibition by a small molecule inhibitor (PRMT5i) in HTLV-1-infected lymphocytes resulted in increased viral gene expression and decreased cellular proliferation. PRMT5i also had selective toxicity in HTLV-1-transformed T-cells. Finally, we demonstrated that PRMT5 and the HTLV-1 p30 protein had an additive inhibitory effect on HTLV-1 gene expression. Our study provides evidence for PRMT5 as a host cell factor important in HTLV-1-mediated T-cell transformation, and a potential target for ATLL treatment. Our next steps are to determine the therapeutic effects of PRMT5i in an in vivo ATL mouse model by examining viral replication kinetics, tumor size, and survival. These studies are the necessary first step to identify PRMT5 as an important epigenetic factor during HTLV-1-mediated cellular transformation and pathogenesis, and will ultimately provide important insight towards its potential as an ATL therapy target.
Research Support

    "Retrovirus Models of Cancer"
    Principle Investigator: Patrick L. Green, Ph.D.
    Agency: National Institutes of Health/National Cancer Institute
    Type: Projects 1-3 Cores A-C of PO1CA100730
    Period: 06/01/20 - 05/31/25

    "Cancer Center Support Grant"
    Principle Investigator: Raphael Pollock, M.D.
    Agency: National Institutes of Health/National Cancer Institute
    Type: P30 CA016058. The overall goal of CCSG is to reduce cancer morbidity and mortality through continued basic, translational and clinical research.
    Role: Associate Director for Basic Research

 

Professional Activities

Awards, Honors, and University Memberships

  • Dale McFarlin Award (outstanding service to the International Retrovirology Association)
  • Robert H. Rainier Chair in Industrial Veterinary Medicine and Research
  • Inaugural David Derse Memorial Lecture/Award
  • Distinguished Alumni Hall of Fame, Xavier High School
  • Ohio State University Distinguished Scholar
  • Fellow, American Academy of Microbiology
  • Fellow, American Association for the Advancement of Science
  • International Retrovirology Association Award for Exceptional Contribution to the Field of HTLV and Related Viruses
  • Leukemia Society of America Scholar
  • Leukemia Society of America Fellow
  • American Cancer Society National Fellow
  • Charles C. Capen Teaching Excellence Award for Graduate Education
  • Pfizer Award for Research Excellence
  • Dorothy and Charles Inbusch Award for Medical Research Excellence
  • Director, Center for Retrovirus Research, The Ohio State University
  • Program Leader, Comprehensive Cancer Center Viral Oncology
  • Joint Appointment, Department of Cancer Genetics
  • Member, Comprehensive Cancer Center
  • Member, Comparative Veterinary Medicine Graduate Program
  • Member, Molecular, Cellular & Developmental Biology Graduate Program
  • Member, Ohio State Biochemistry Graduate Program
  • Member, Integrated Biomedical Sciences Graduate Program

Professional Service

  • Editor, AIDS Research and Human Retroviruses
  • Member, Editorial Board of Retrovirology
  • Member, Editorial Board of The Open Virology Journal
  • Member, Editorial Board of Virology: Research and Treatment
  • Treasurer, International Retrovirology Association
  • Reviewer, NIH Intramural and Extramural Research Programs

Graduate Training Record

  • Past: 11 post-doctoral fellows, 19 PhD students, 1 medical student, all are employed as assistant/associate professors, staff in industry or government agencies, research scientists, or postdoctoral fellows
  • Past: 4 junior faculty
  • Past: 4 undergraduate students
  • Past: 9 DVM students
  • Currently: 3 junior faculty,  1 post-doctoral fellow, 7 DVM students

Publications

Peer Reviewed Articles

(partial listing, 2019 - present)

  • Tu JJ, King E, Maksimova V, Smith S, Macias R, Cheng X, Yu L, Ratner L, Green PL, Niewiesk S, Richner J, Panfil AR. An HTLV-1 envelope mRNA vaccine is immunogenic and protective in New Zealand rabbits. J Virol. 2024 Jan 9:e0162323. doi: 10.1128/jvi.01623-23. Epub ahead of print.
  • Smith S, Seth J, Midkiff A, Stahl R, Syu Y-C, Shkriabai N, Kvaratskhelia M, Musier-Forsyth K, Jain P, Green PL, Panfil AR. The pleiotropic effects of YBX1 on HTLV-1 transcription. Int J Mol Sci. 2023 Aug 23;24(17):13119. doi: 10.3390/ijms241713119. PMID: 37685922; PMCID: PMC10487795.
  • Polakowski N, Sarker MAK, Hoang K, Boateng G, Rushing A, Kendle W, Pique C, Green PL, Panfil AR, Lemasson I. HBZ upregulates myoferlin expression to facilitate HTLV-1 Infection. PLoS Pathog. 2023 Feb 24;19(2):e1011202. doi: 10.1371/journal.ppat.1011202. eCollection 2023 Feb. PMID: 36827461
  • Ernzen K, Melvin C, Yu L, Phelps C, Niewiesk S, Green PL, Panfil AR.  The PRMT5 inhibitor EPZ015666 selectively targets HTLV-1-transformed T-cell lines in vitro and in vivo. Front. Microbiol. Volume 14 - 2023 | https://doi.org/10.3389/fmicb.2023.1101544.
  • Poondru S, Joseph A, Harding JC, Sundaramoorthi H, Mehta-Shah N, Green PL, Hassan A, Rauch DA, Ratner L. Adult T-cell leukemia-lymphoma presenting concurrently with myelopathy.  Case Rep Oncol 2022;15:918–926. DOI:10.1159/000525174.  PMID: 36636671 PMCID: PMC9830276
  • Maksimova V, Smith S, Seth J, Phelps C, Niewiesk S, Satou Y, Green PL, Panfil AR. HTLV-1 intragenic viral enhancer influences immortalization phenotype in vitro, but is dispensable for persistence and disease development in animal models. Front Immunol. 2022 Jul 25;13:954077 DOI:10.3389/fimmu.2022.954077.
  • Moore RM, Green PL, Fingland RB, Hare T.  Impactful research improving animal health through a one health approach. Am J Vet Res 2022. Vol 83:Issue 8. https://doi.org/10.2460/ajvr.22.06.0103
  • Daenthanasanmak A, Bamford RN, Yoshioka M, Yang S-M, Homan P, Karim B, Bryant BR, Petrus MN, Thomas CJ, Green PL, Miljkovic MD, Conlon KC, Waldmann TA.  Triple combination of BET plus PI3K and NF-κB inhibitors exhibit synergistic activity in adult T cell leukemia/lymphoma.  Blood Adv 2022 Apr 12;6(7):2346-2360. doi: 10.1182/bloodadvances.2021005948 PMID: 35030628
  • Ishio T, Kumar S, Shimono J, Daenthanasanmak A, Dubois S, Lin Y, Bryant BR, Petrus MN, Bachy E, Huang DW, Yang Y, Green PL, Hasegawa H, Maeda M, Goto H, Endo T, Yokota T, Hatanaka KC, Hatanaka Y, Tanaka S, Matsuno Y, Yang Y, Hashino S, Teshima T, Waldmann TA, Staudt LM, Nakagawa M. Genome-wide CRISPR screen identifies CDK6 as a therapeutic target in Adult T-cell leukemia/lymphoma. Blood. 2022 Mar 10;139(10):1541-1556. doi: 10.1182/blood.2021012734 PMID:34818414
  • Cheng X, Joseph A, Castro V, Chen-Liaw A, Skidmore Z, Ueno T, Fujisawa JI, Rauch DA, Challen GA, Martinez MP, Green PL, Griffith M, Payton JE, Edwards JR, Ratner L. Epigenomic regulation of human T cell leukemia virus by chromatin-insulator CTCF. PLoS Pathog. 2021 May 21;17(5):e1009577. doi:10.1371/journal.ppat.1009577. eCollection 2021 May. PMID:34019588 PMCID: PMC8174705
  • Panfil AR, Green PL, Yoder KE. CRISPR genome editing applied to the pathogenic retrovirus HTLV-1. Front Cell Infect Microb. 2020 Dec23;10:580371.doi:10.3389/fcimb.2020.580371.  PMCID: PMC7785941
  • Panfil AR, Green PL.  HTLV-1 and HTLV-2.   In: Encyclopedia of Virology Fourth Edition. Bramford D and Zuckerman M, editors. Volume 2, 2021, Pages 528-539. Elsevier.  https://doi.org/10.1016/B978-0-12-809633-8.21225-9.
  • Martinez MP, Cheng X, Ancy JJ, Al-Saleem J, Panfil A, Palettas M, Dirksen WP, Ratner L, Green PL. HTLV-1 CTCF-Binding Site is Dispensable for In Vitro Immortalization and Persistent Infection In Vivo. Retrovirology, 2019 Dec 21;16(1):44. doi: 10.1186/s12977-019-0507-9. PMCID:PMC6925871.
  • 1Xiang J, Huey D, Rauch DA, Panfil A, Cheng X, Harding JC, Su X, Fontana F, Xu Y, Esser A, Wong W-H, Jia J, Sundaramoorthi H, Wu K, Rosol TJ, Veis D, Green PL, Ratner L, Niewiesk S, Weilbaecher KN. HTLV-1 Viral Oncogene HBZ Drives Bone Destruction in Adult T Cell Leukemia. JCI Insight. 2019 Oct 3;4(19). pii: 128713. doi: 10.1172/jci.insight.128713. PMCID: PMC6795409.
  • Kohart NA, Elshafae SM, Supsahvad W, Alasonyalilar-Demirer A, Panfil AR, Xiang J, Dirksen WP, Veis DJ, Green PL, Weilbaecher KN, Rosol TJ. Mouse Model Recapitulates the Phenotypic Heterogeneity of Human Adult T-cell Leukemia/Lymphoma in Bone.  J Bone Oncology 2019 Aug 20;19:100257. doi: 10.1016/j.jbo.2019.100257. eCollection 2019 Dec. PMCID:PMC6911918.
  • 1Martinez MP, Al-Saleem JJ, Green PL: Comparative Virology of HTLV-1 and HTLV-2. Retrovirology, 2019 Aug 7;16(1):21. doi: 10.1186/s12977-019-0483-0. PMCID: PMC6686503.
  • Al-Saleem JJ, Kvaratskhelia M, Ratner L, Green PL: Identification and Characterization of a novel Tax-1  interacting protein, SNX27, and its role in HTLV-1 pathobiology. PLoS One 2019 Mar 21;14(3):e0214059. doi: 10.1371/journal.pone.0214059. eCollection 2019. PMCID: PMC6428263.