? PROJECT 2 Adult T-cell leukemia/lymphoma (ATL) develops in a subset of people infected with HTLV-1 and is an aggressive T-cell malignancy. ATL?s unique relationship to bone (long latency in the marrow, bone invasion, osteolytic lesions, and hypercalcemia) makes it an ideal model to dissect the critical factors that support tumor development and progression in bone. Thus, our work on ATL will likely shed light on other late recurring and bone-tropic tumors like multiple myeloma, breast, and prostate cancer. Using transgenic mice, we showed that the HTLV-1 tax viral oncogene can mediate both ATL development as well as osteolytic bone destruction through effects on bone-resorbing osteoclasts (OCs). However, Tax expression is downregulated in ~70% of human ATL, despite ongoing bone involvement and bone loss, suggesting that another viral factor is important. Recently, we found that HBZ, a second HTLV-1 oncogene, can also lead to lymphoproliferative disease and pathologic bone loss when expressed transgenically or in a humanized mouse model of HTLV-1 infection. RANKL plays an important role in this process, but is not a direct target of HBZ. We and others find that HBZ upregulates the expression of Wnt5a and heparanase (HPSE), tumor-derived paracrine factors that modulate the tumor microenvironment in bone and are upregulated in patient ATL cells. Wnt5a activates noncanonical Wnt signaling via Ror2 and has both osteoblast-inhibiting and OC-stimulating activities, including increasing RANKL expression. HPSE enzymatically cleaves heparan sulfate, thereby altering cell surfaces and extracellular matrix, increasing bioavailability of growth factors and cytokines including RANKL and likely Wnt5a. We hypothesize that hbz expression in transformed ATL cells reprograms the bone microenvironment via increasing Wnt5a and Heparanase expression, and thereby affects tumor progression and bone loss. Our plan for evaluating this hypothesis relies integrally on the in vivo models that were developed during the previous funding period, including new patient-derived xenograft (PDX) models that cause systemic bone loss in mice following intraperitoneal (IP) implantation and newly characterized local bone effects of established ATL cell lines following implantation into bone (with intratibial injection; IT). We will make extensive use of viral vectors to manipulate Wnt5a and HPSE in these ATL and PDX lines, primarily with CRISPR/Cas9, taking advantage of the expertise of Dr. Yoder (Vector Core 1). In collaboration with Dr. Niewiesk (Animal Core 2), we have also implemented a humanized immune system (HIS) model in which immunodeficient mice are transplanted with human cord blood cells, and then infected with HTLV-1, modeling emergence of lymphoproliferative disease (LPD) accompanied by systemic bone loss. This collaboration will be a key part of our experimental design.

Public Health Relevance

? PROJECT 2 Adult T-cell leukemia/lymphoma (ATL), an aggressive leukemia that is associated with hypercalcemia and osteolytic bone lesions develops in 2-4% of HTLV-1 infected individuals. We will investigate new roles for two bone-acting factors, regulated by the HTLV-1 viral oncogene Hbz, in reprogramming the bone environment. These represent therapeutic opportunities to target the significant bone loss in ATL patients, as well as affect progression of ATL tumors which are often resistant to standard chemotherapy.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Program Projects (P01)
Project #
2P01CA100730-16A1
Application #
10023352
Study Section
Special Emphasis Panel (ZCA1)
Project Start
2003-04-21
Project End
2025-05-31
Budget Start
2020-07-01
Budget End
2021-06-30
Support Year
16
Fiscal Year
2020
Total Cost
Indirect Cost
Name
Ohio State University
Department
Type
DUNS #
832127323
City
Columbus
State
OH
Country
United States
Zip Code
43210
Murali, Bhavna; Ren, Qihao; Luo, Xianmin et al. (2018) Inhibition of the Stromal p38MAPK/MK2 Pathway Limits Breast Cancer Metastases and Chemotherapy-Induced Bone Loss. Cancer Res 78:5618-5630
Huey, Devra D; Niewiesk, Stefan (2018) Production of Humanized Mice through Stem Cell Transfer. Curr Protoc Mouse Biol 8:17-27
Romeo, Megan; Hutchison, Tetiana; Malu, Aditi et al. (2018) The human T-cell leukemia virus type-1 p30II protein activates p53 and induces the TIGAR and suppresses oncogene-induced oxidative stress during viral carcinogenesis. Virology 518:103-115
Cherian, Mathew A; Olson, Sydney; Sundaramoorthi, Hemalatha et al. (2018) An activating mutation of interferon regulatory factor 4 (IRF4) in adult T-cell leukemia. J Biol Chem 293:6844-6858
Huey, Devra D; Bolon, Brad; La Perle, Krista M D et al. (2018) Role of Wild-type and Recombinant Human T-cell Leukemia Viruses in Lymphoproliferative Disease in Humanized NSG Mice. Comp Med 68:4-14
Pérès, Eléonore; Blin, Juliana; Ricci, Emiliano P et al. (2018) PDZ domain-binding motif of Tax sustains T-cell proliferation in HTLV-1-infected humanized mice. PLoS Pathog 14:e1006933
Panfil, Amanda R; Al-Saleem, Jacob; Howard, Cory M et al. (2018) Stability of the HTLV-1 Antisense-Derived Protein, HBZ, Is Regulated by the E3 Ubiquitin-Protein Ligase, UBR5. Front Microbiol 9:80
Kenney, Adam D; Dowdle, James A; Bozzacco, Leonia et al. (2017) Human Genetic Determinants of Viral Diseases. Annu Rev Genet 51:241-263
Webb, Lindsay M; Amici, Stephanie A; Jablonski, Kyle A et al. (2017) PRMT5-Selective Inhibitors Suppress Inflammatory T Cell Responses and Experimental Autoimmune Encephalomyelitis. J Immunol 198:1439-1451
Singh, Gatikrushna; Fritz, Sarah M; Ranji, Arnaz et al. (2017) Isolation of Cognate RNA-protein Complexes from Cells Using Oligonucleotide-directed Elution. J Vis Exp :

Showing the most recent 10 out of 162 publications