Abstract

Abstract Initially studied mainly for its role as a regulator of neural cell differentiation and development, expression of HDAC11 was once thought to be restricted exclusively to brain, kidney and testes. Hence, our recent discovery that HDAC11 acts as an important modulator of antigen presentation and T cell activation, downregulating IL-10 transcription via interactions with the IL-10 promoter at the chromatin level, exposes a previously unknown capacity and tissue specificity for this enzyme. Transgenic mice harboring an eGFP reporter construct driven by the HDAC11 promoter (Tg-HDAC11-eGFP) (Heinz, N Nat. Rev. Neuroscience 2001) clearly illustrate the dynamic changes in HDAC11 gene expression in hematopoietic cell lineages, additionally unveiling an important role for HDAC11 in B cell lymphopoiesis and plasma cell biology. While common lymphoid progenitors appear to be devoid of HDAC11 transcriptional activation as indicated by minimal detectable eGFP expression, eGFP intensity markedly increases in the B-1 stage of differentiation in the periphery. Interestingly, examination of both the bone marrow (BM) and peripheral blood (PB) plasma cell compartment demonstrates an increase in expression of eGFP/HDAC11 mRNA at the steady-state, and these results are consistent with HDAC11 expression measured in PB from healthy human subjects. Furthermore, mice globally deficient in HDAC11 expression (HDAC11KO mice) exhibit a 50% decrease in plasma cells in both the bone marrow and peripheral blood plasma cell compartments relative to wild-type mice. The concordance of HDAC11 expression and plasma cell differentiation leads us to hypothesize that HDAC11 may also be critical to malignant plasma cell survival. A comparison of normal bone marrow and malignant plasma cells isolated from multiple myeloma patient samples reveals a significantly higher level of HDAC11 expression associated with malignancy. Similar results are observed in 8 of 12 myeloma cell lines suggesting that HDAC11 expression may provide a distinct survival advantage to malignant plasma cells. Further stratification of patients into “newly diagnosed” and “proteasome inhibitor resistant” categories defines a positive correlation between HDAC11 expression and refractory disease. Treatment of the myeloma cell lines with Quisinostat, a second-generation HDAC inhibitor with enhanced selectivity for HDAC 1, 2, 4, 10 and 11 induces growth retardation at low nanomolar concentrations. Future studies will entail direct targeting of HDAC11 in myeloma cell lines and patient specimens to determine the contribution of HDAC11 to Quinsinostat activity. Taken together, we have unveiled a previously unknown role for HDAC11 in plasma cell differentiation and survival. The demonstration of HDAC11 overexpression in primary human myeloma cells provides a framework for therapeutics targeting this HDAC in multiple myeloma. Citation Format: Eva Sahakian, Jason Brayer, John Powers, Mark Meads, Susan Deng, Allison Distler, Melissa Alsina, Taiga Nishihori, Rachid Baz, Alejandro Villagra, Javier Pinilla-Ibarz, Eduardo Sotomayor, Kenneth Shain. Histone deacetylase 11 (HDAC11) regulates B cell lymphopoiesis and potentiates plasma cell survival in multiple myeloma. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 5537. doi:10.1158/1538-7445.AM2014-5537

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call