Upregulation of Focal Adhesion Kinase, a Potential Therapeutic Target, in Acute Myeloid Leukemia (AML) and Myelodysplastic Syndromes (MDS).

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2827-2827
Author(s):  
Hui Yang ◽  
Carlos E. Bueso-Ramos ◽  
Sherry A. Pierce ◽  
Yue Wei ◽  
Zhihong Fang ◽  
...  

Abstract Abstract 2827 Introduction The myelodysplastic syndromes (MDS) are a group of leukemia characterized by bone marrow failure and increased risk of transformation to acute myelogenous leukemia (AML). Focal adhesion kinase (FAK), a non-receptor protein tyrosine kinase (PTK), plays a key role in the integration of protein-mediated signal transduction. It is a critical mediator of the integrin signaling cascade, which modulates cell proliferation, apoptosis, adhesion, spreading and migration. FAK is upregulated in a wide variety of human cancers. It has been reported that expression of FAK in acute myeloid leukemia (AML) is associated with enhanced blast migration, increased cellularity and poor prognosis. Furthermore, FAK silencing inhibited leukemogenesis in BCR-ABL-transformed hematopoietic cells. FAK has been proposed a potential target in cancer therapy. In this study, we studied the expression patterns of FAK in 98 patients with MDS and performed preclinical studies of FAK inhibition in leukemia cell lines. Methods CD34+ cells from 98 newly diagnosed patients with MDS and 5 normal donor bone marrow specimens were sorted with CD34 isolation kit from Miltenyi Biotec. Total cellular RNA was isolated using Trizol, cDNA was obtained by using TaqMan reverse transcription reagent (Applied Biosystems). For real-time PCR, FAK assay were purchased from Applied Biosystems and analyzed with an Applied Biosystems Prism 7500 Sequencing detection system. GAPDH was used as internal control. Immunohistochemistry was used to detect FAK protein level in cytospin from MDS CD34+ cells. FAK antibody was obtained from abcam. FAK inhibitor was purchased from Santa Cruz Biotechnology. Results For the 98 MDS patients studied, 78% were older than 60 years; by IPSS score, 17 (17.3%) low risk, 35 (35.7%) INT-1, 24 (24.5%) INT-2, 10 (10.2%) high risk, 12 (12.2%) not available. By cytogenetics, diploid 44 (45%), 20q- 7 (7%), -5/5q- 7 (7%), -7/7q- 7 (7%), -5/5q- and -7/7q- 6 (6%), 8+ 6 (6%), IM 6 (6%), MISC 14 (14%). By real-time PCR, we observed FAK overexpression in 28% of MDS CD34+ cells (fold 2.2–26). We then analyzed FAK protein expression in 10 MDS CD34+ cell cytospin with either higher or lower RNA expression by QPCR using immunohistochemistry. The protein expression patterns were 100% consistent with RNA expression level. This result suggests that FAK expression is upregulated in MDS CD34+ cells. We then performed analysis of clinical associations between FAK expression and clinical characteristics. No association was observed in particular between FAK expression and survival. Because of the potential role of FAK as a therapeutic target, we then studied the effects of FAK inhibition in cell lines. We studied FAK expression level in leukemia cell lines of AML origin and found high protein expression level of FAK in AML leukemia cell line OCI-AML3 and KG1. We treated these cells with FAK inhibitor 14 and detected a dose dependent anti-proliferative effect on both cell lines. Using Annexin V - FITC analysis by flow cytometry, we found the FAK inhibition could induce apoptosis in both cell lines at concentrations of 10uM both at 24 hours and 48 hours. Conclusions This study shows that FAK is aberrantly expressed in MDS CD34+ cells, together with the anti-proliferative and apoptosis induction effect of FAK inhibitor, our study demonstrates that FAK may be a potential therapeutic target in MDS and AML patients. Disclosures: No relevant conflicts of interest to declare.

Glycobiology ◽  
2015 ◽  
Vol 26 (4) ◽  
pp. 343-352 ◽  
Author(s):  
Silvina Laura Lompardía ◽  
Mariángeles Díaz ◽  
Daniela Laura Papademetrio ◽  
Marilina Mascaró ◽  
Matías Pibuel ◽  
...  

2000 ◽  
Vol 24 (9) ◽  
pp. 741-749 ◽  
Author(s):  
Kimo C Stine ◽  
Bryce A Warren ◽  
Robert L Saylors ◽  
David L Becton

Blood ◽  
1996 ◽  
Vol 88 (9) ◽  
pp. 3383-3390 ◽  
Author(s):  
AM Turner ◽  
NL Lin ◽  
S Issarachai ◽  
SD Lyman ◽  
VC Broudy

FLT3 ligand is a hematopoietic growth factor that plays a key role in growth of primitive hematopoietic cells. FLT3 receptor mRNA is found in early hematopoietic progenitors and in human myeloid leukemia blasts. Much less is known about the surface expression of FLT3 receptor on human hematopoietic cells. Using human 125I-FLT3 ligand, we have identified and characterized surface FLT3 receptors on normal and malignant human hematopoietic cells and cell lines. Our results showed that surface display of FLT3 receptor was greatest in fresh myeloid leukemia blast cells and myeloid leukemia cell lines. Erythroleukemic and megakaryocytic leukemia cell lines (n = 5) bound little to no 125I-FLT3 ligand. Scatchard analysis of 125I-FLT3 ligand binding data shows that three myeloid leukemia cell lines, ML-1, AML-193, and HL-60, as well as normal human marrow mononuclear cells, exhibit high affinity FLT3 receptors. Crosslinking of 125I-FLT3 ligand to FLT3 receptors on the surface of ML-1 myeloid leukemia cells indicates that the FLT3 ligand. The rates of FLT3 ligand internalization and degradation were determined by binding 125I-FLT3 ligand to ML-1 cells and acid stripping to distinguish surface bound from internalized ligand. Internalized 125I-FLT3 ligand was detected within 5 minutes after binding to ML-1 cells. In addition, we evaluated the effect of FLT3 ligand on megakaryocytic colony growth and nuclear endoreduplication, alone or in the presence of thrombopoietin. FLT3 ligand did not promote colony forming unit megakaryocyte (CFU-Meg) colony growth or megakaryocyte nuclear maturation, nor did FLT3 ligand augment the effects of thrombopoietin on these measures of megakaryopoiesis. These data indicate that the FLT3 receptor shares several characteristics with the c-kit receptor including dimerization and rapid internalization. However, the more restricted cellular distribution of the FLT3 receptor may target the effects of FLT3 ligand to primitive hematopoietic cells and to myeloid and lymphoid progenitor cells, in contrast to the pleiotropic effects of the c-kit receptor ligand, stem cell factor.


Blood ◽  
1998 ◽  
Vol 92 (5) ◽  
pp. 1497-1504 ◽  
Author(s):  
Zhu-Gang Wang ◽  
Roberta Rivi ◽  
Laurent Delva ◽  
Andrea König ◽  
David A. Scheinberg ◽  
...  

Abstract Inorganic arsenic trioxide (As2O3) and the organic arsenical, melarsoprol, were recently shown to inhibit growth and induce apoptosis in NB4 acute promyelocytic leukemia (APL) and chronic B-cell leukemia cell lines, respectively. As2O3 has been proposed to principally target PML and PML-RAR proteins in APL cells. We investigated the activity of As2O3 and melarsoprol in a broader context encompassing various myeloid leukemia cell lines, including the APL cell line NB4-306 (a retinoic acid–resistant cell line derived from NB4 that no longer expresses the intact PML-RAR fusion protein), HL60, KG-1, and the myelomonocytic cell line U937. To examine the role of PML in mediating arsenical activity, we also tested these agents using murine embryonic fibroblasts (MEFs) and bone marrow (BM) progenitors in which the PML gene had been inactivated by homologous recombination. Unexpectedly, we found that both compounds inhibited cell growth, induced apoptosis, and downregulated bcl-2 protein in all cell lines tested. Melarsoprol was more potent than As2O3 at equimolar concentrations ranging from 10−7 to 10−5 mol/L. As2O3 relocalized PML and PML-RAR onto nuclear bodies, which was followed by PML degradation in NB4 as well as in HL60 and U937 cell lines. Although melarsoprol was more potent in inhibiting growth and inducing apoptosis, it did not affect PML and/or PML-RAR nuclear localization. Moreover, both As2O3 and melarsoprol comparably inhibited growth and induced apoptosis of PML+/+ and PML−/− MEFs, and inhibited colony-forming unit erythroid (CFU-E) and CFU granulocyte-monocyte formation in BM cultures of PML+/+ and PML−/− progenitors. Together, these results show that As2O3 and melarsoprol inhibit growth and induce apoptosis independent of both PML and PML-RAR expression in a variety of myeloid leukemia cell lines, and suggest that these agents may be more broadly used for treatment of leukemias other than APL. © 1998 by The American Society of Hematology.


2007 ◽  
Vol 31 (4) ◽  
pp. 497-506 ◽  
Author(s):  
Tobias Berg ◽  
Yalin Guo ◽  
Mahmoud Abdelkarim ◽  
Manfred Fliegauf ◽  
Michael Lübbert

2011 ◽  
Vol 52 (11) ◽  
pp. 2139-2147 ◽  
Author(s):  
Carine Tang ◽  
Lisa Schafranek ◽  
Dale B. Watkins ◽  
Wendy T. Parker ◽  
Sarah Moore ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document