Dendritic cells from CML patients have altered actin organization, reduced antigen processing, and impaired migration

R Dong, K Cwynarski, A Entwistle… - Blood, The Journal …, 2003 - ashpublications.org
R Dong, K Cwynarski, A Entwistle, F Marelli-Berg, F Dazzi, E Simpson, JM Goldman…
Blood, The Journal of the American Society of Hematology, 2003ashpublications.org
Chronic myeloid leukemia (CML) is characterized by expression of the BCR-ABL fusion
gene that encodes a 210-kDa protein, which is a constitutively active tyrosine kinase. At
least 70% of the oncoprotein is localized to the cytoskeleton, and several of the most
prominent tyrosine kinase substrates for p210BCR-ABLare cytoskeletal proteins. Dendritic
cells (DCs) are bone marrow–derived antigen-presenting cells responsible for the initiation
of immune responses. In CML patients, up to 98% of myeloid DCs generated from peripheral …
Chronic myeloid leukemia (CML) is characterized by expression of the BCR-ABL fusion gene that encodes a 210-kDa protein, which is a constitutively active tyrosine kinase. At least 70% of the oncoprotein is localized to the cytoskeleton, and several of the most prominent tyrosine kinase substrates for p210BCR-ABLare cytoskeletal proteins. Dendritic cells (DCs) are bone marrow–derived antigen-presenting cells responsible for the initiation of immune responses. In CML patients, up to 98% of myeloid DCs generated from peripheral blood mononuclear cells areBCR-ABL positive. In this study we have compared the morphology and behavior of myeloid DCs derived from CML patients with control DCs from healthy individuals. We show that the actin cytoskeleton and shape of CML-DCs of myeloid origin adherent to fibronectin differ significantly from those of normal DCs. CML-DCs are also defective in processing and presentation of exogenous antigens such as tetanous toxoid. The antigen-processing defect may be a consequence of the reduced capacity of CML-DCs to capture antigen via macropinocytosis or via mannose receptors when compared with DCs generated from healthy individuals. Furthermore, chemokine-induced migration of CML-DCs in vitro was significantly reduced. These observations cannot be explained by a difference in the maturation status of CML and normal DCs, because phenotypic analysis by flow cytometry showed a similar surface expression of maturation makers. Taken together, these results suggest that the defects in antigen processing and migration we have observed in CML-DCs may be related to underlying cytoskeletal changes induced by the p210BCR-ABLfusion protein.
ashpublications.org