4.7 Article

TPL2 kinase regulates the inflammatory milieu of the myeloma niche

Journal

BLOOD
Volume 123, Issue 21, Pages 3305-3315

Publisher

AMER SOC HEMATOLOGY
DOI: 10.1182/blood-2014-02-554071

Keywords

-

Categories

Funding

  1. International Myeloma Foundation
  2. Kirschstein National Research Service Award from National Human Genome Research Institute [T32HL007899]
  3. training grant from National Human Genome Research Institute [T32HG002760]
  4. Wisconsin Alumni Research Foundation through the University of Wisconsin (UW) Graduate School
  5. TL1 trainee award from the Clinical and Translational Science Awards program through the National Center for Research Resources grant [1UL1RR025011]
  6. National Center for Advancing Translational Sciences [9U54TR000021]
  7. UW Carbone Cancer Center Trillium Fund for Multiple Myeloma Research
  8. UW Department of Medicine
  9. UW Carbone Cancer Center [P30 CA014520]
  10. UW-Madison School of Medicine and Public Health

Ask authors/readers for more resources

Targeted modulation of microenvironmental regulatory pathways may be essential to control myeloma and other genetically/clonally heterogeneous cancers. Here we report that human myeloma-associated monocytes/macrophages (MAM), but not myeloma plasma cells, constitute the predominant source of interleukin-1 beta (IL-1 beta), IL-10, and tumor necrosis factor-alpha at diagnosis, whereas IL-6 originates from stromal cells and macrophages. To dissect MAM activation/cytokine pathways, we analyzed Toll-like receptor (TLR) expression in human myeloma CD14(+) cells. We observed coregulation of TLR2 and TLR6 expression correlating with local processing of versican, a proteoglycan TLR2/6 agonist linked to carcinoma progression. Versican has not been mechanistically implicated in myeloma pathogenesis. We hypothesized that the most readily accessible target in the versican-TLR2/6 pathway would be the mitogen-activated protein 3 (MAP3) kinase, TPL2 (Cot/MAP3K8). Ablation of Tpl2 in the genetically engineered in vivo myeloma model, V kappa*MYC, led to prolonged disease latency associated with plasma cell growth defect. Tpl2 loss abrogated the inflammatory switch in MAM within nascent myeloma lesions and licensed macrophage repolarization in established tumors. MYC activation/expression in plasma cells was independent of Tpl2 activity. Pharmacologic TPL2 inhibition in human monocytes led to dose-dependent attenuation of IL-1 beta induction/secretion in response to TLR2 stimulation. Our results highlight a TLR2/6-dependent TPL2 pathway as novel therapeutic target acting nonautonomously through macrophages to control myeloma progression.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available