The Platelet-Derived Growth Factor Receptor beta Fuses to Two Distinct Loci at 3p21 in Imatinib Responsive Chronic Eosinophilic Leukemia.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3253-3253 ◽  
Author(s):  
Claire Curtis ◽  
Jane F. Apperley ◽  
Raymond Dang ◽  
Michael Jeng ◽  
Jason Gotlib ◽  
...  

Abstract We have identified three patients (2 adults, one infant) who presented with BCR-ABL negative eosinophilic myeloproliferative disorders. Cytogenetic analysis revealed a t(1;3;5)(p36;p21;q33) for case 1 and a t(3;5)(p21–25;q31–35) for cases 2 and 3. Two-color fluorescence in situ hybridization (FISH) using differentially labelled probes flanking PDGFRB indicated that this gene was disrupted in all three cases. 5′ rapid amplification of cDNA ends (5′RACE) for case 1 identified an in-frame mRNA fusion of exon 9 of the WDR48 gene at 3p21 to exon 12 of PDGFRB. The chimeric mRNA is predicted to encode a 872 amino acid fusion protein that retains the amino terminal WD repeat region of WDR48 fused to the transmembrane and intracellular tyrosine kinase domains of PDGFRbeta. Cases 2 and 3 were negative for the WDR48-PDGFRB fusion mRNA by RT-PCR using several combinations of primers. 5′RACE PCR from case 2 RNA identified a fusion involving a second 3p21 gene: GOLGA4 exon 11 was fused in-frame to exon 11 of PDGFRB. Exactly the same fusion was found in case 3. The predicted 991 amino acid protein included the amino terminal coiled-coil domain of GOLGA4 fused to the transmembrane and intracellular tyrosine kinase domains of PDGFRbeta. Interestingly, both WDR48 and GOLGA4 are involved in endocytic shuttling pathways. The presence of all fusions was confirmed by RT-PCR and identification of the genomic breakpoints. Imatinib, a known inhibitor of PDGFRbeta, selectively blocked the growth of patient CFU-GM for case 2. Following the identification of PDGFRB rearrangements, all three patients were treated with imatinib. Case 1 was in transformation, but responded rapidly to minimal doses of imatinib (800mg daily for 4 days) with complete cytogenetic remission but remained pancytopenic. Blast crisis recurred 8 months later, responded similarly to 3 days of imatinib, but the patient died 2 months later of invasive fungal infection. Case 2 responded clinically and remains in sustained cytogenetic and molecular remission (nested RT-PCR negative for GOLGA4-PDGFRB). Case 3 (a 13 month old boy) had a complete hematologic response to 50mg/day imatinib but the t(3;5) was still seen in 40% of metaphases at 3 months. We conclude that PDGFRB fuses to diverse partner genes to give rise to atypical MPDs. Although very rare, identification of these fusions is critical for proper management of affected individuals.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 667-667
Author(s):  
Christoph Walz ◽  
Georgia Metzgeroth ◽  
Claudia Schoch ◽  
Torsten Haferlach ◽  
Rudiger Hehlmann ◽  
...  

Abstract Fusion genes involving PDGFRA, PDGFRB, FGFR1 and JAK2 are seen in a substantial number of patients with BCR-ABL negative myeloproliferative disorders (MPD) and result in constitutive activation of the corresponding tyrosine kinase moiety. The vast majority of tyrosine kinase fusion partners contain coiled-coil domains or other dimerization motifs properties that are essential for malignant transformation. We have identified two patients presenting with eosinophilia-associated MPD and a t(5;12)(q31;q24) or a complex translocation t(1;5;11) with involvement of 5q31, respectively, suggesting a possible involvement of the PDGFRB gene which is located at chromosome band 5q31–33. 5′-rapid amplification of cDNA ends (5′-RACE) for the t(5;12) identified an in-frame mRNA fusion between ’G protein-coupled receptor kinase interactor 2′ (GIT2) exon 12 at 12q24 and PDGFRB exon 11. GIT2 is a member of the GIT protein family that is extensively alternative spliced in many distinct forms causing its functional diversity. A reciprocal transcript was amplified by RT-PCR with a fusion between PDGFRB exon 10 and GIT2 exon 13. GIT2-PDGFRB is predicted to be translated into a 742 amino acid fusion protein that retains the GIT2 N-terminal protein-protein interaction motif Ankyrin and an Arf GTPase activating protein (ArfGAP) domain fused to the transmembrane and catalytic domain of PDGFRB. The truncated GIT2 protein lacks coiled-coil domains as they are lost in the fusion protein due to the breakpoint within GIT2 intron 12. We therefore speculate that the Ankyrin repeat, which is one of the most common protein-protein interaction motifs in nature, may have replaced the function of a coiled-coil domain offering dimerization properties to the fusion protein. 5′-RACE for the complex t(1;5;11) identified an in-frame mRNA fusion between ’GPI-anchored membrane protein 1′ (GPIAP1) exon 7 at 11p13 and PDGFRB exon 11. Normal GPIAP1 is a cytoplasmic phosphoprotein which plays a mainly uncharacterized role in cellular activation or proliferation. The chimeric mRNA is predicted to encode an 803 amino acid fusion protein retaining the coiled-coil domain of GPIAP1 fused to the transmembrane and catalytic domains of PDGFRB. Both patients have been treated with 400 mg/day imatinib, which is a selective inhibitor of PDGFRB, and achieved rapid complete clinical and hematological remission. Residual GIT2-PDGFRB transcripts could be detected repeatedly during a 17 months follow up in case 1 whereas no follow-up samples have been available for case 2. These data give further evidence that numerous partner genes fuse to PDGFRB in BCR-ABL negative MPDs. In addition, the data demonstrate that cytogenetic analysis is a mandatory technique for the identification of tyrosine kinase fusion genes. In cases with abnormalities of chromosome 5q, a possible involvement of PDGFRB should be screened by adequate FISH and PCR-based techniques. Although their occurrence is rare in general, the identification of these fusion genes is essential for the successful treatment with tyrosine kinase inhibitors.


2020 ◽  
Author(s):  
Jens Schittenhelm ◽  
Lukas Ziegler ◽  
Jan Sperveslage ◽  
Michel Mittelbronn ◽  
David Capper ◽  
...  

Abstract Background Fibroblast growth factor receptor (FGFR) inhibitors are currently used in clinical development. A subset of glioblastomas carries gene fusion of FGFR3 and transforming acidic coiled-coil protein 3. The prevalence of other FGFR3 alterations in glioma is currently unclear. Methods We performed RT-PCR in 101 glioblastoma samples to detect FGFR3-TACC3 fusions (“RT-PCR cohort”) and correlated results with FGFR3 immunohistochemistry (IHC). Further, we applied FGFR3 IHC in 552 tissue microarray glioma samples (“TMA cohort”) and validated these results in two external cohorts with 319 patients. Gene panel sequencing was carried out in 88 samples (“NGS cohort”) to identify other possible FGFR3 alterations. Molecular modeling was performed on newly detected mutations. Results In the “RT-PCR cohort,” we identified FGFR3-TACC3 fusions in 2/101 glioblastomas. Positive IHC staining was observed in 73/1024 tumor samples of which 10 were strongly positive. In the “NGS cohort,” we identified FGFR3 fusions in 9/88 cases, FGFR3 amplification in 2/88 cases, and FGFR3 gene mutations in 7/88 cases in targeted sequencing. All FGFR3 fusions and amplifications and a novel FGFR3 K649R missense mutation were associated with FGFR3 overexpression (sensitivity and specificity of 93% and 95%, respectively, at cutoff IHC score > 7). Modeling of these data indicated that Tyr647, a residue phosphorylated as a part of FGFR3 activation, is affected by the K649R mutation. Conclusions FGFR3 IHC is a useful screening tool for the detection of FGFR3 alterations and could be included in the workflow for isocitrate dehydrogenase (IDH) wild-type glioma diagnostics. Samples with positive FGFR3 staining could then be selected for NGS-based diagnostic tools.


1991 ◽  
Vol 11 (6) ◽  
pp. 3191-3202 ◽  
Author(s):  
O Segatto ◽  
F Lonardo ◽  
D Wexler ◽  
F Fazioli ◽  
J H Pierce ◽  
...  

The epidermal growth factor receptor (EGFR) and gp185erbB-2 are closely related tyrosine kinases. Despite extensive sequence and structural homology, these two receptors display quantitative and qualitative differences in their ability to couple with mitogenic signalling pathways. By using chimeric molecules between EGFR and erbB-2, we found that the determinants responsible for the specificity of mitogenic signal transduction are located in the amino-terminal half of the tyrosine kinase domain of either receptor. In the EGFR, mutational analysis within this subdomain revealed that deletion of residues 660 to 667 impaired receptor mitogenic activity without affecting its tyrosine kinase properties. This sequence is therefore likely to contribute to the specificity of substrate recognition by the EGFR kinase.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 687-687
Author(s):  
Daniela Cilloni ◽  
Sonia Carturan ◽  
Enrico Bracco ◽  
Ilaria Defilippi ◽  
Chiara Maffè ◽  
...  

Abstract The abnormal activation of tyrosine kinases are a common finding in chronic myeloproliferative disorders. Perturbation of RTK signalling by genetic alterations results into an abnormal proliferation advantage and finally into a malignant transformation. c-Ros is an orphan RTK displaying transforming activity whose role has been established in the development of neuronal neoplasia. The aim of this study was to evaluate the involvement of c-Ros in the pathogenesis of chronic myelomonocytic leukemia (CMML) and to establish the effects of c-Ros activation. c-Ros expression was evaluated by RQ-PCR in 133 samples collected from 96 CMML patients at diagnosis (96 BM and 37 PB) and 60 healthy donors (30 PB and 30 BM). The protein amount and localization was analyzed by westen blot and immunofluorescence assay. In order to establish the effects of c-Ros activation we generated a chimeric receptor containing the extracellular domain derived from epidermal growth factor receptor (EGFR) and the transmembrane and cytoplasmic domains from c-ros (ER). The chimeric receptor was then transfected in NIH3T3 and HEK293T cells. Transfected and control cells were then stimulated with 100 nM EGF ligand and proliferation and apoptosis evaluated by incorporation of 3H timidine and MTT assay and by FACS for the detection of annexin V, respectively. We found that Ros is undetectable in healthy subjects but it is overexpressed in CMML (p<0,0001) in both BM and PB cells with a median value of 2−ΔΔCt in BM of 380 (range 10–63303) and 212 in PB (range 6–30012). WB confirmed the presence of c-Ros protein in CMML cells but not in normal controls. Immunofluorescence staining localized the protein within the cytoplasm. We found that ROS is highly expressed in CD34+ cells and monocytes from CMML patients but not in their normal counterparts. Sequence analysis revealed the absence of mutations of c-Ros promoter. SNPs analysis exclude the presence of duplications or deletions of the gene. Moreover we found that the EGF induced activation of c-Ros affects proliferation by increasing of 3.5 folds the proliferation rate as compared to cells transfected with the empty vector and stimulated with EGF under the same conditions. Furthermore cell adhesion was 4 folds decreased as compared to control. By contrast apoptosis is not affected by c-ROS activity (p=0,2). This study demonstrates that c-Ros is abnormally expressed in patients with CMML. The abnormal activation of c-Ros is responsible for loss of adhesion and increased proliferation. In conclusion, we identified a new tyrosine kinase which may be responsible for the proliferation defect typical of CMML cells and could represent a target for molecular therapies.


2000 ◽  
Vol 113 (11) ◽  
pp. 1963-1971 ◽  
Author(s):  
R.J. Finst ◽  
P.J. Kim ◽  
E.R. Griffis ◽  
L.M. Quarmby

A key event in deflagellation or deciliation is the severing of the nine outer-doublet axonemal microtubules at a specific site in the flagellar transition zone. Previous genetic analysis revealed three genes that are essential for deflagellation in Chlamydomonas. We have now identified the first of these products, Fa1p, a protein required for Ca(2+)-dependent, axonemal microtubule severing. Genetic mapping and the availability of a tagged allele allowed us to physically map the gene to the centromere-proximal domain of the mating-type locus. We identified clones of Chlamydomonas genomic DNA that rescued the Ca(2+)-dependent axonemal microtubule severing defect of fa1 mutants. The FA1 cDNA, obtained by RT-PCR, encodes a novel protein of 171 kDa, which is predicted to contain an amino-terminal coiled-coil domain and three Ca(2+)/calmodulin binding domains. By western analysis and subcellular fractionation, the FA1 product is enriched in flagellar-basal body complexes. Based on these observations and previous studies, we hypothesize that a Ca(2+)-activated, Ca(2+)-binding protein binds Fa1p leading ultimately to the activation of axonemal microtubule severing.


Blood ◽  
2003 ◽  
Vol 101 (4) ◽  
pp. 1520-1529 ◽  
Author(s):  
Jonathan J. Keats ◽  
Tony Reiman ◽  
Christopher A. Maxwell ◽  
Brian J. Taylor ◽  
Loree M. Larratt ◽  
...  

This study analyzed the frequency and clinical significance of t(4;14)(p16;q32) in multiple myeloma (MM) among 208 patients with MM and 52 patients with monoclonal gammopathy of undetermined significance (MGUS); diagnosed between 1994 and 2001. Patients with the translocation were identified using reverse transcription–polymerase chain reaction (RT-PCR) to detect hybrid immunoglobulin heavy chain (IgH)–MMSET transcripts from the der(4) chromosome. We found 31 (14.9%) t(4;14)+ MM patients and 1 (1.9%) t(4;14)+ MGUS patient. IgH-MMSET hybrid transcripts were detected in bone marrow (BM) and blood. Breakpoint analysis revealed that 67.7% of t(4;14)+ patients expressed hybrid transcripts potentially encoding full-length MMSET, whereas the remainder lacked one or more amino terminal exons. Expression of fibroblast growth factor receptor 3 (FGFR3), presumptively dysregulated on der(14), was detected by RT-PCR in only 23 of 31 (74%) patients with t(4;14)+ MM. Patients lacking FGFR3 expression also lacked detectable der(14) products. Longitudinal analysis of 53 MM patients with multiple BM and blood samples showed that, over time, BM from t(4;14)+ patients remained positive and that t(4;14)− patients did not acquire the translocation. IgH-MMSET hybrid transcripts and FGFR3 transcripts disappeared from blood during response to therapy. No correlation was observed between the occurrence of t(4;14) and known prognostic indicators. However, we find the t(4;14) translocation predicts for poor survival (P = .006; median, 644 days vs 1288 days; hazard ratio [HR], 2.0), even in FGFR3 nonexpressors (P = .003). The presence of t(4;14) is also predictive of poor response to first-line chemotherapy (P = .05). These results indicate a significant clinical impact of the t(4;14) translocation in MM that is independent of FGFR3 expression.


1989 ◽  
Vol 9 (1) ◽  
pp. 24-33 ◽  
Author(s):  
D Martin-Zanca ◽  
R Oskam ◽  
G Mitra ◽  
T Copeland ◽  
M Barbacid

Molecular analysis of the human trk oncogene, a transforming gene isolated from a colon carcinoma biopsy, revealed the existence of a novel member of the tyrosine kinase gene family. This locus, which we now designate the trk proto-oncogene, codes for a protein of 790 amino acid residues that has several features characteristic of cell surface receptors. They include (i) a 32-amino-acid-long putative signal peptide, (ii) an amino-terminal moiety (residues 33 to 407) rich in consensus sites for N-glycosylation, (iii) a transmembrane domain, (iv) a kinase catalytic region highly related to that of other tyrosine kinases, and (v) a very short (15 residue) carboxy-terminal tail. Residues 1 to 392 were absent in the trk oncogene, as they were replaced by tropomyosin sequences. However, no other differences were found between the transforming and nontransforming trk alleles (residues 392 to 790), suggesting that no additional mutations are required to activate the transforming potential of this gene. The human trk proto-oncogene codes for a 140,000-dalton glycoprotein, designated gp140proto-trk. However, its primary translational product is a 110,000-dalton glycoprotein which becomes immediately glycosylated, presumably during its translocation into the endoplasmic reticulum. This molecule, designated gp110proto-trk, is further glycosylated to yield the mature form, gp140proto-trk. Both gp110proto-trk and gp140proto-trk proteins possess in vitro kinase activity specific for tyrosine residues. Finally, iodination of intact NIH 3T3 cells expressing trk proto-oncogene products indicated that only the mature form, gp140proto-trk, cross the plasma membrane, becoming exposed to the outside of the cell. These results indicate that the product of the human trk locus is a novel tyrosine kinase cell surface receptor for an as yet unknown ligand.


1991 ◽  
Vol 11 (6) ◽  
pp. 3191-3202
Author(s):  
O Segatto ◽  
F Lonardo ◽  
D Wexler ◽  
F Fazioli ◽  
J H Pierce ◽  
...  

The epidermal growth factor receptor (EGFR) and gp185erbB-2 are closely related tyrosine kinases. Despite extensive sequence and structural homology, these two receptors display quantitative and qualitative differences in their ability to couple with mitogenic signalling pathways. By using chimeric molecules between EGFR and erbB-2, we found that the determinants responsible for the specificity of mitogenic signal transduction are located in the amino-terminal half of the tyrosine kinase domain of either receptor. In the EGFR, mutational analysis within this subdomain revealed that deletion of residues 660 to 667 impaired receptor mitogenic activity without affecting its tyrosine kinase properties. This sequence is therefore likely to contribute to the specificity of substrate recognition by the EGFR kinase.


1995 ◽  
Vol 108 (7) ◽  
pp. 2683-2692
Author(s):  
J. Marshall ◽  
D.V. Holberton

Previously described extended proteins from the cytoskeleton of Giardia lamblia (beta-giardin, median body protein) have been found to be segmented coiled coils with regular structural repeat patterns in their amino acid sequences. Screening a lambda ZAPII library derived from Giardia genomic DNA with an antibody directed against a 34 × 10(3) M(r) giardin isoform selected a gene encoding a much larger polypeptide chain (HPSR2), the sequence of which was determined by chromosome walking the open reading frame. The complete gene has been cloned and expressed as a recombinant protein of 183 × 10(3) M(r). The predicted amino acid sequence of the protein has identifiable features suggesting that it might be a motor protein with an amino-terminal hydrolytic domain attached to a long coiled coil stalk. The presumed head domain is 211 residues and contains a P-loop sequence conserved in purine nucleotide-binding proteins. The remaining 1409 amino acids mainly make up a region of heptad repeats such as in myosin or the kinesin stalk, ending in a small (67 amino acids) carboxy-terminal domain. Fourier analysis of the predicted stalk shows the presence of a strong physical repeat created by regular heptad phase changes dividing the coil into segments of 25 residues. This structure most closely resembles the smaller microtubule-associated median body protein which has segments of 24 residues.


Blood ◽  
2003 ◽  
Vol 102 (12) ◽  
pp. 4187-4190 ◽  
Author(s):  
Kathryn Wilkinson ◽  
Elvira R. P. Velloso ◽  
Luiz Fernando Lopes ◽  
Charles Lee ◽  
Jon C. Aster ◽  
...  

Abstract Eosinophilia is common in myeloproliferative disorders (MPDs) with abnormalities of chromosome band 5q31-33, including those that present with t(1;5)(q23;q33). With the development of rational drug therapy, characterization of the molecular targets for these translocations could guide treatment and affect patient survival. We cloned the t(1;5)(q23;q33) and showed that it fuses platelet-derived growth factor receptor beta (PDGFRB) to the coiled-coil domains of a novel partner protein, myomegalin. Using two-color interphase fluorescence in situ hybridization (FISH), we also demonstrated that the eosinophils are clonal in these disorders. Imatinib mesylate has recently been shown to be efficacious in MPDs with PDGFR activation. Therefore, following our molecular studies, we were able to redirect this patient's treatment. Although she had refractory and progressive disease, once imatinib was started, complete clinical and hematologic remission, as well as major cytogenetic response, was achieved. Given the therapeutic implications, our findings stress the need to aggressively investigate the molecular basis of these diseases, with emphasis on the PDGFR family. (Blood. 2003;102: 4187-4190)


Sign in / Sign up

Export Citation Format

Share Document