Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Proc Natl Acad Sci U S A ; 117(32): 19221-19227, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32719139

ABSTRACT

Despite the outstanding success of the cancer drug imatinib, one obstacle in prolonged treatment is the emergence of resistance mutations within the kinase domain of its target, Abl. We noticed that many patient-resistance mutations occur in the dynamic hot spots recently identified to be responsible for imatinib's high selectivity toward Abl. In this study, we provide an experimental analysis of the mechanism underlying drug resistance for three major resistance mutations (G250E, Y253F, and F317L). Our data settle controversies, revealing unexpected resistance mechanisms. The mutations alter the energy landscape of Abl in complex ways: increased kinase activity, altered affinity, and cooperativity for the substrates, and, surprisingly, only a modestly decreased imatinib affinity. Only under cellular adenosine triphosphate (ATP) concentrations, these changes cumulate in an order of magnitude increase in imatinib's half-maximal inhibitory concentration (IC50). These results highlight the importance of characterizing energy landscapes of targets and its changes by drug binding and by resistance mutations developed by patients.


Subject(s)
Antineoplastic Agents/pharmacology , Imatinib Mesylate/pharmacology , Neoplasms/enzymology , Oncogene Proteins v-abl/genetics , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Drug Resistance, Neoplasm , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Oncogene Proteins v-abl/chemistry , Oncogene Proteins v-abl/metabolism
2.
Biochem J ; 474(23): 3963-3984, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29025973

ABSTRACT

Discoidin, CUB, and LCCL domain containing 2 (DCBLD2) is a neuropilin-like transmembrane scaffolding receptor with known and anticipated roles in vascular remodeling and neuronal positioning. DCBLD2 is also up-regulated in several cancers and can drive glioblastomas downstream of activated epidermal growth factor receptor. While a few studies have shown either a positive or negative role for DCBLD2 in regulating growth factor receptor signaling, little is known about the conserved signaling features of DCBLD family members that drive their molecular activities. We previously identified DCBLD2 tyrosine phosphorylation sites in intracellular YxxP motifs that are required for the phosphorylation-dependent binding of the signaling adaptors CRK and CRKL (CT10 regulator of kinase and CRK-like). These intracellular YxxP motifs are highly conserved across vertebrates and between DCBLD family members. Here, we demonstrate that, as for DCBLD2, DCBLD1 YxxP motifs are required for CRKL-SH2 (Src homology 2) binding. We report that Src family kinases (SFKs) and Abl differentially promote the interaction between the CRKL-SH2 domain and DCBLD1 and DCBLD2, and while SFKs and Abl each promote DCBLD1 and DCBLD2 binding to the CRKL-SH2 domain, the effect of Abl is more pronounced for DCBLD1. Using high-performance liquid chromatography coupled with tandem mass spectrometry, we quantified phosphorylation at several YxxP sites in DCBLD1 and DCBLD2, mapping site-specific preferences for SFKs and Abl. Together, these data provide a platform to decipher the signaling mechanisms by which these novel receptors drive their biological activities.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Membrane Proteins/chemistry , Nuclear Proteins/chemistry , Oncogene Proteins v-abl/chemistry , Proto-Oncogene Proteins c-fyn/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cloning, Molecular , Conserved Sequence , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Proteins v-abl/metabolism , Phosphorylation , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Zebrafish
3.
FASEB J ; 29(9): 3750-61, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25999467

ABSTRACT

RAS proteins are signal transduction gatekeepers that mediate cell growth, survival, and differentiation through interactions with multiple effector proteins. The RAS effector RAS- and RAB-interacting protein 1 (RIN1) activates its own downstream effectors, the small GTPase RAB5 and the tyrosine kinase Abelson tyrosine-protein kinase (ABL), to modulate endocytosis and cytoskeleton remodeling. To identify ABL substrates downstream of RAS-to-RIN1 signaling, we examined human HEK293T cells overexpressing components of this pathway. Proteomic analysis revealed several novel phosphotyrosine peptides, including Harvey rat sarcoma oncogene (HRAS)-pTyr(137). Here we report that ABL phosphorylates tyrosine 137 of H-, K-, and NRAS. Increased RIN1 levels enhanced HRAS-Tyr(137) phosphorylation by nearly 5-fold, suggesting that RAS-stimulated RIN1 can drive ABL-mediated RAS modification in a feedback circuit. Tyr(137) is well conserved among RAS orthologs and is part of a transprotein H-bond network. Crystal structures of HRAS(Y137F) and HRAS(Y137E) revealed conformation changes radiating from the mutated residue. Although consistent with Tyr(137) participation in allosteric control of HRAS function, the mutations did not alter intrinsic GTP hydrolysis rates in vitro. HRAS-Tyr(137) phosphorylation enhanced HRAS signaling capacity in cells, however, as reflected by a 4-fold increase in the association of phosphorylated HRAS(G12V) with its effector protein RAF proto-oncogene serine/threonine protein kinase 1 (RAF1). These data suggest that RAS phosphorylation at Tyr(137) allosterically alters protein conformation and effector binding, providing a mechanism for effector-initiated modulation of RAS signaling.


Subject(s)
Oncogene Proteins v-abl/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/physiology , Amino Acid Substitution , Animals , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mutation, Missense , Oncogene Proteins v-abl/chemistry , Oncogene Proteins v-abl/genetics , Phosphorylation/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/genetics , Rats , Tyrosine/chemistry , Tyrosine/genetics , Tyrosine/metabolism , rab5 GTP-Binding Proteins/chemistry , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism , raf Kinases/chemistry , raf Kinases/genetics , raf Kinases/metabolism
4.
Eur J Haematol ; 86(5): 361-71, 2011 May.
Article in English | MEDLINE | ID: mdl-21435002

ABSTRACT

Chromosomal rearrangements involving the ABL1 gene, leading to a BCR-ABL1 fusion gene, have been mainly associated with chronic myeloid leukemia and B-cell acute lymphoblastic leukemia (ALL). At present, six other genes have been shown to fuse to ABL1. The kinase domain of ABL1 is retained in all chimeric proteins that are also composed of the N-terminal part of the partner protein that often includes a coiled-coil or a helix-loop-helix domain. These latter domains allow oligomerization of the protein that is required for tyrosine kinase activation, cytoskeletal localization, and neoplastic transformation. Fusion genes that have a break in intron 1 or 2 (BCR-ABL1, ETV6-ABL1, ZMIZ1-ABL1, EML1-ABL1, and NUP214-ABL1) have transforming activity, although NUP214-ABL1 requires amplification to be efficient. The NUP214-ABL1 gene is the second most prevalent fusion gene involving ABL1 in malignant hemopathies, with a frequency of 5% in T-cell ALL. Both fusion genes (SFPQ-ABL1 and RCSD1-ABL1) characterized by a break in intron 4 of ABL1 are associated with B-cell ALL, as the chimeric proteins lacked the SH2 domain of ABL1. Screening for ABL1 chimeric genes could be performed in patients with ALL, more particularly in those with T-cell ALL because ABL1 modulates T-cell development and plays a role in cytoskeletal remodeling processes in T cells.


Subject(s)
Genes, abl , Hematologic Neoplasms/genetics , Oncogene Fusion , Cell Transformation, Neoplastic/genetics , Hematologic Neoplasms/chemistry , Humans , Intracellular Signaling Peptides and Proteins/genetics , Oncogene Proteins v-abl/antagonists & inhibitors , Oncogene Proteins v-abl/chemistry , Oncogene Proteins v-abl/genetics , Oncogene Proteins, Fusion/genetics , PTB-Associated Splicing Factor , Protein-Tyrosine Kinases/genetics , RNA-Binding Proteins/genetics , Transcription Factors/genetics
5.
J Chem Theory Comput ; 16(12): 7852-7865, 2020 Dec 08.
Article in English | MEDLINE | ID: mdl-33147951

ABSTRACT

Imatinib, a drug used for the treatment of chronic myeloid leukemia and other cancers, works by blocking the catalytic site of pathological constitutively active Abl kinase. While the binding pose is known from X-ray crystallography, the different steps leading to the formation of the complex are not well understood. The results from extensive molecular dynamics simulations show that imatinib can primarily exit the known crystallographic binding pose through the cleft of the binding site or by sliding under the αC helix. Once displaced from the crystallographic binding pose, imatinib becomes trapped in intermediate states. These intermediates are characterized by a high diversity of ligand orientations and conformations, and relaxation timescales within this region may exceed 3-4 ms. Analysis indicates that the metastable intermediate states should be spectroscopically indistinguishable from the crystallographic binding pose, in agreement with tryptophan stopped-flow fluorescence experiments.


Subject(s)
Imatinib Mesylate/chemistry , Molecular Dynamics Simulation , Oncogene Proteins v-abl/chemistry , Protein Kinase Inhibitors/chemistry , Binding Sites/drug effects , Crystallography, X-Ray , Humans , Imatinib Mesylate/pharmacology , Oncogene Proteins v-abl/antagonists & inhibitors , Oncogene Proteins v-abl/metabolism , Protein Kinase Inhibitors/pharmacology
6.
J Virol ; 82(11): 5307-15, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18367522

ABSTRACT

Abelson murine leukemia virus (Ab-MLV) arose from a recombination between gag sequences in Moloney MLV (Mo-MLV) and the c-abl proto-oncogene. The v-Abl oncoprotein encoded by Ab-MLV contains MA, p12, and a portion of CA sequences derived from the gag gene of Mo-MLV. Previous studies indicated that alteration of MA sequences affects the biology of Mo-MLV and Ab-MLV. To understand the role of these sequences in Ab-MLV transformation more fully, alanine substitution mutants that affect Mo-MLV replication were examined in the context of Ab-MLV. Mutations affecting Mo-MLV replication decreased transformation, while alanine mutations in residues dispensable for Mo-MLV replication did not. The altered v-Abl proteins displayed aberrant subcellular localization that correlated to transformation defects. Immunofluorescent analyses suggested that aberrant trafficking of the altered proteins and improper interaction with components of the cytoskeleton were involved in the phenotype. Similar defects in localization were observed when the Gag moiety containing these mutations was expressed in the absence of abl-derived sequences. These results indicate that MA sequences within the Gag moiety of the v-Abl protein contribute to proper localization by playing a dominant role in trafficking of the v-Abl molecule.


Subject(s)
Abelson murine leukemia virus/metabolism , Gene Products, gag/chemistry , Gene Products, gag/metabolism , Moloney murine leukemia virus/metabolism , Oncogene Proteins v-abl/chemistry , Oncogene Proteins v-abl/metabolism , Abelson murine leukemia virus/chemistry , Abelson murine leukemia virus/genetics , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Dimerization , Gene Products, gag/genetics , Models, Molecular , Molecular Sequence Data , Moloney murine leukemia virus/chemistry , Moloney murine leukemia virus/genetics , Mutation/genetics , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Oncogene Proteins v-abl/genetics , Peptides/chemistry , Peptides/metabolism , Protein Structure, Quaternary , Protein Structure, Tertiary , Sequence Alignment , Sequence Homology, Amino Acid
7.
Anal Biochem ; 385(2): 300-8, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19059193

ABSTRACT

Conformational change is a common molecular mechanism for the regulation of kinase activities. Small molecule modulators of protein conformations, including allosteric kinase inhibitors, are highly wanted as tools for the interrogation of kinase biology and as selective therapeutic agents. However, straightforward cellular assays monitoring kinase conformations in a manner conducive to high-throughput screening (HTS) are not readily available. Here we describe such an HTS-compatible conformational sensor assay for Abl based on a split luciferase construct. The Abl sensor responds to intramolecular structural rearrangements associated with intracellular Abl deactivation and small molecule inhibition. The intact regulatory CAP-SH3-SH2 domain is required for the full functionality of the sensor. Moreover, a T334I Abl mutant (T315I in Abl1a) was found to be particularly well suited for HTS purposes and mechanistic intracellular studies of T334I mutant inhibitors. We expect that the split luciferase-based conformational sensor approach might be more broadly useful to probe the intracellular activation of other kinases and enzymes in general.


Subject(s)
Mutation, Missense , Oncogene Proteins v-abl/analysis , Animals , Cell Line , Enzyme Inhibitors/pharmacology , Humans , Luciferases , Oncogene Proteins v-abl/chemistry , Oncogene Proteins v-abl/genetics , Protein Conformation/drug effects
8.
Mol Cell Biol ; 18(11): 6795-804, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9774693

ABSTRACT

In Abelson murine leukemia virus (A-MuLV)-transformed cells, members of the Janus kinase (Jak) family of non-receptor tyrosine kinases and the signal transducers and activators of transcription (STAT) family of signaling proteins are constitutively activated. In these cells, the v-Abl oncoprotein and the Jak proteins physically associate. To define the molecular mechanism of constitutive Jak-STAT signaling in these cells, the functional significance of the v-Abl-Jak association was examined. Mapping the Jak1 interaction domain in v-Abl demonstrates that amino acids 858 to 1080 within the carboxyl-terminal region of v-Abl bind Jak1 through a direct interaction. A mutant of v-Abl lacking this region exhibits a significant defect in Jak1 binding in vivo, fails to activate Jak1 and STAT proteins, and does not support either the proliferation or the survival of BAF/3 cells in the absence of cytokine. Cells expressing this v-Abl mutant show extended latency and decreased frequency in generating tumors in nude mice. In addition, inducible expression of a kinase-inactive mutant of Jak1 protein inhibits the ability of v-Abl to activate STATs and to induce cytokine-independent proliferation, indicating that an active Jak1 is required for these v-Abl-induced signaling pathways in vivo. We propose that Jak1 is a mediator of v-Abl-induced STAT activation and v-Abl induced proliferation in BAF/3 cells, and may be important for efficient transformation of immature B cells by the v-abl oncogene.


Subject(s)
Cell Division/physiology , Interleukin-3/physiology , Oncogene Proteins v-abl/chemistry , Protein-Tyrosine Kinases/metabolism , Animals , Binding Sites/physiology , Cell Line , Gene Expression Regulation/genetics , Janus Kinase 1 , Mice , Mice, Nude , Neoplasms, Experimental , Protein Binding , Signal Transduction/physiology , Transcriptional Activation/physiology , Transfection/genetics
9.
Mol Cell Biol ; 20(2): 628-33, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10611241

ABSTRACT

Abelson murine leukemia virus (A-MuLV) is an acute transforming retrovirus that preferentially transforms early B-lineage cells both in vivo and in vitro. Its transforming protein, v-Abl, is a tyrosine kinase related to v-Src but containing an extended C-terminal domain. Many mutations affecting the C-terminal portion of the molecule block the pre-B-transforming activity of v-Abl without affecting the fibroblast-transforming ability. In this study we have determined the abilities of both wild-type and C-terminally truncated (p90) forms of v-Abl to transform cells from p53(-/-) mice. Lack of p53 increases the susceptibility of bone marrow cells to transformation by v-Abl by a factor of more than 7 but does not alter v-Abl's preference for B220(+) IgM(-) pre-B cells. p53-deficient mice have earlier tumor onset, more rapid tumor progression, and decreased survival time following A-MuLV infection, but all of the tumors are pre-B lymphomas. Thus, p53-dependent pathways inhibit v-Abl transformation but play no role in conferring preferential transformation of pre-B cells. Surprisingly, the C-terminally truncated form of v-Abl (p90) transforms pre-B cells very efficiently in mice lacking p53, thus demonstrating that the C terminus of v-Abl does not determine preB tropism but is necessary to overcome p53-dependent inhibition of transformation.


Subject(s)
B-Lymphocytes/pathology , Cell Transformation, Neoplastic/pathology , Gene Deletion , Lymphoma/pathology , Oncogene Proteins v-abl/metabolism , Tumor Suppressor Protein p53/physiology , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Cell Lineage , Cell Transformation, Neoplastic/genetics , Cell Transformation, Viral , Cells, Cultured , Disease Progression , Female , Genetic Predisposition to Disease/genetics , Genotype , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/virology , Leukemia Virus, Murine/enzymology , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/pathogenicity , Lymphoma/genetics , Lymphoma/mortality , Lymphoma/virology , Male , Mice , Mice, Knockout , Oncogene Proteins v-abl/chemistry , Oncogene Proteins v-abl/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Tumor Stem Cell Assay , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
10.
Protein Sci ; 25(8): 1378-84, 2016 08.
Article in English | MEDLINE | ID: mdl-27241634

ABSTRACT

Understanding the conformational propensities of proteins is key to solving many problems in structural biology and biophysics. The co-variation of pairs of mutations contained in multiple sequence alignments of protein families can be used to build a Potts Hamiltonian model of the sequence patterns which accurately predicts structural contacts. This observation paves the way to develop deeper connections between evolutionary fitness landscapes of entire protein families and the corresponding free energy landscapes which determine the conformational propensities of individual proteins. Using statistical energies determined from the Potts model and an alignment of 2896 PDB structures, we predict the propensity for particular kinase family proteins to assume a "DFG-out" conformation implicated in the susceptibility of some kinases to type-II inhibitors, and validate the predictions by comparison with the observed structural propensities of the corresponding proteins and experimental binding affinity data. We decompose the statistical energies to investigate which interactions contribute the most to the conformational preference for particular sequences and the corresponding proteins. We find that interactions involving the activation loop and the C-helix and HRD motif are primarily responsible for stabilizing the DFG-in state. This work illustrates how structural free energy landscapes and fitness landscapes of proteins can be used in an integrated way, and in the context of kinase family proteins, can potentially impact therapeutic design strategies.


Subject(s)
Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Oncogene Proteins v-abl/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Amino Acid Motifs , Databases, Protein , Humans , Kinetics , Ligands , Mitogen-Activated Protein Kinase 14/chemistry , Models, Molecular , Oncogene Proteins v-abl/chemistry , Protein Binding , Protein Domains , Protein Structure, Secondary , Structural Homology, Protein , Thermodynamics
11.
Oncogene ; 18(37): 5138-47, 1999 Sep 16.
Article in English | MEDLINE | ID: mdl-10498863

ABSTRACT

Human and mouse Abelson interacting proteins (Abi) are SH3-domain containing proteins that bind to the proline-rich motifs of the Abelson protein tyrosine kinase. We report a new member of this gene family, a Drosophila Abi (dAbi) that is a substrate for Abl kinase and that co-immunoprecipitates with Abl if the Abi SH3 domain is intact. We have identified a new function for both dAbi and human Abi-2 (hAbi-2). Both proteins activate the kinase activity of Abl as assayed by phosphorylation of the Drosophila Enabled (Ena) protein. Removal of the dAbi SH3 domain eliminates dAbi's activation of Abl kinase activity. dAbi is an unstable protein in cells and is present at low steady state levels but its protein level is increased coincident with phosphorylation by Abl kinase. Expression of the antisense strand of dAbi reduces dAbi protein levels and abolishes activation of Abl kinase activity. Modulation of Abi protein levels may be an important mechanism for regulating the level of Abl kinase activity in the cell.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/physiology , Cytoskeletal Proteins , Drosophila Proteins , Drosophila melanogaster/enzymology , Homeodomain Proteins/physiology , Insect Proteins/isolation & purification , Oncogene Proteins v-abl/physiology , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins c-abl/physiology , Abelson murine leukemia virus/enzymology , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Carrier Proteins/isolation & purification , Cloning, Molecular , Consensus Sequence , Drosophila melanogaster/genetics , Enzyme Activation , Evolution, Molecular , Homeodomain Proteins/chemistry , Humans , Insect Proteins/chemistry , Insect Proteins/genetics , Insect Proteins/physiology , Mice , Molecular Sequence Data , Oncogene Proteins v-abl/chemistry , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Protein-Tyrosine Kinases/chemistry , Proto-Oncogene Proteins c-abl/chemistry , Rats , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity , Transfection , src Homology Domains
12.
Oncogene ; 19(21): 2523-31, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10851051

ABSTRACT

The Abl oncoproteins v-Abl and BCR-Abl can activate member of the signal transducers and activators of transcription (STAT) family of signaling proteins. The mechanisms by which these oncoproteins activate STATs appear to differ. In cells transformed by v-Abl, Janus kinase (JAK) tyrosine kinases are constitutively activated. In these cells, the v-Abl oncoprotein and the JAK kinases physically associate. Mapping of the JAK interaction domain in v-Abl demonstrates that amino acids within the carboxyl terminal region of v-Abl bind JAKs through a direct interaction. A mutant of v-Abl lacking this region does not bind or activate JAK 1 in vivo, fails to activate STAT proteins, does not induce cellular proliferation, and is less efficient in cellular transformation. Kinase inactive mutants of JAK 1 inhibit the ability of v-Abl to activate STATs, to induce cytokine-independent proliferation, and to transform bone marrow cells. Interestingly, these effects correlate with defects in the activation of several pathways by v-Abl including Akt, PI3-kinase, STATs, and Ras. These data suggest that Jak kinases may play an important role in v-Abl induced transformation. Oncogene (2000).


Subject(s)
DNA-Binding Proteins/metabolism , Genes, abl/physiology , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Trans-Activators/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/enzymology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Enzyme Activation , Genes, abl/genetics , Humans , Janus Kinase 1 , Oncogene Proteins v-abl/chemistry , Oncogene Proteins v-abl/genetics , Oncogene Proteins v-abl/metabolism , Protein-Tyrosine Kinases/chemistry , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , STAT1 Transcription Factor
13.
Leukemia ; 18(4): 864-71, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14973502

ABSTRACT

Mutations of the ABL kinase domain (KD) are common in patients with chronic myelogenous leukemia (CML) who develop resistance to imatinib. We developed an RT-PCR-based denaturing high-performance liquid chromatography (D-HPLC) assay to detect mutations of the ABL KD. Validation experiments using mixtures of wild type and mutant amplicons showed that the D-HPLC assay could detect mutant transcripts when they represented at least 15% of the total, and was thus twice as sensitive as automated sequencing. When D-HPLC was applied to 30 cDNAs from patients with imatinib resistance that had previously been characterized for KD mutations by direct sequencing of BCR-ABL RT-PCR products, there was concordance in 97% of samples. Resequencing confirmed the original mutations in all cases. In addition, sequencing of individual clones detected a mutation in one sample that had been mutation-positive by D-HPLC but wild type by conventional sequencing. In serial samples from the same individuals, D-HPLC detected mutations as early as 260 days before hematological relapse. D-HPLC is suitable for routine clinical monitoring of CML patients for emergence of KD mutations and may be useful for optimizing therapy. Early detection of emerging mutant clones may aid in guiding decisions regarding alternative treatment options.


Subject(s)
Chromatography, High Pressure Liquid/methods , DNA Mutational Analysis/methods , Oncogene Proteins v-abl/genetics , Adult , Aged , Benzamides , Chromatography, High Pressure Liquid/standards , DNA Mutational Analysis/standards , DNA, Neoplasm/genetics , Drug Resistance/genetics , Female , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Male , Middle Aged , Oncogene Proteins v-abl/chemistry , Piperazines/therapeutic use , Protein Structure, Tertiary , Pyrimidines/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity
14.
Science ; 347(6224): 882-6, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25700521

ABSTRACT

Macromolecular function is rooted in energy landscapes, where sequence determines not a single structure but an ensemble of conformations. Hence, evolution modifies a protein's function by altering its energy landscape. Here, we recreate the evolutionary pathway between two modern human oncogenes, Src and Abl, by reconstructing their common ancestors. Our evolutionary reconstruction combined with x-ray structures of the common ancestor and pre-steady-state kinetics reveals a detailed atomistic mechanism for selectivity of the successful cancer drug Gleevec. Gleevec affinity is gained during the evolutionary trajectory toward Abl and lost toward Src, primarily by shifting an induced-fit equilibrium that is also disrupted in the clinical T315I resistance mutation. This work reveals the mechanism of Gleevec specificity while offering insights into how energy landscapes evolve.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Drug Resistance, Neoplasm/genetics , Evolution, Molecular , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , src-Family Kinases/chemistry , Antineoplastic Agents/chemistry , Benzamides/chemistry , Entropy , Humans , Imatinib Mesylate , Mutation , Oncogene Proteins v-abl/chemistry , Oncogene Proteins v-abl/genetics , Phylogeny , Piperazines/chemistry , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Structure, Secondary , Pyrimidines/chemistry , src-Family Kinases/classification , src-Family Kinases/genetics
15.
Leuk Lymphoma ; 30(3-4): 225-35, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9713955

ABSTRACT

Following the discovery of the p210bcrabl protein product of the bcrabl chimeric fusion gene generated by the Philadelphia chromosome translocation in chronic myelogenous leukemia (CML), structure function studies quickly identified which parts of this molecule were playing a role in the generation of the phenotypes of growth factor independent growth, anchorage independent growth, and genetic instability which are associated with this disease. These latter changes result in abnormally high levels of mature myeloid elements circulating in the systemic circulation of CML patients. In addition, the genetic instability which is associated with the presence of the Philadelphia chromosome drives the evolution of the disease from an indolent chronic non life-threatening leukemia, to a fulminant acute leukemic syndrome which results in the death of patients from bleeding and infection. Multiple sites of contact between the p210bcrabl and its substrates have already been identified which are relevant to the phenotypic changes characteristic of CML cells and define their response to therapy. In this review, we will discuss what is known about the relationships between the structural domains of the p210bcrabl protein and the characteristics of the disease process which it causes. We will also discuss how this information may be applied to the establishment of new directions in therapy.


Subject(s)
Fusion Proteins, bcr-abl/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mutagens , Oncogene Proteins v-abl/physiology , Cell Differentiation , Cell Division , Cell Transformation, Neoplastic , Fusion Proteins, bcr-abl/chemistry , Humans , Oncogene Proteins v-abl/chemistry
17.
Nat Struct Mol Biol ; 17(4): 519-27, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20357770

ABSTRACT

Interactions between Src homology 2 (SH2) domains and phosphotyrosine sites regulate tyrosine kinase signaling networks. Selective perturbation of these interactions is challenging due to the high homology among the 120 human SH2 domains. Using an improved phage-display selection system, we generated a small antibody mimic (or 'monobody'), termed HA4, that bound to the Abelson (Abl) kinase SH2 domain with low nanomolar affinity. SH2 protein microarray analysis and MS of intracellular HA4 interactors showed HA4's specificity, and a crystal structure revealed how this specificity is achieved. HA4 disrupted intramolecular interactions of Abl involving the SH2 domain and potently activated the kinase in vitro. Within cells, HA4 inhibited processive phosphorylation activity of Abl and also inhibited STAT5 activation. This work provides a design guideline for highly specific and potent inhibitors of a protein interaction domain and shows their utility in mechanistic and cellular investigations.


Subject(s)
Antibodies, Monoclonal/immunology , Oncogene Proteins v-abl/immunology , src Homology Domains , Amino Acid Sequence , Models, Molecular , Molecular Sequence Data , Oncogene Proteins v-abl/chemistry , Phosphorylation , Sequence Homology, Amino Acid
18.
J Virol ; 77(11): 6208-15, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12743277

ABSTRACT

The v-Abl protein encoded by Abelson murine leukemia virus (Ab-MLV) induces transformation of pre-B cells via a two-stage process. An initial proliferative phase during which cells with limited tumorigenic potential expand is followed by a crisis period marked by high levels of apoptosis and erratic growth. Transformants that survive this phase emerge as fully malignant cells and usually contain mutations that disable the p53 tumor suppressor pathway. Consistent with the importance of p53 in this process, pre-B cells from p53 null animals bypass crisis. Thus, the transformation process reflects a balance between signals from the v-Abl protein that drive transformation and those coming from the cellular response to inappropriate growth. One prediction of this hypothesis is that Ab-MLV mutants that are compromised in their ability to transform cells may be less equipped to overcome the effects of p53. To test this idea, we examined the ability of the P120/R273K mutant to transform pre-B cells from wild-type, p53 null, and Ink4a/Arf null mice. The SH2 domain of the v-Abl protein encoded by this mutant contains a substitution that affects the phosphotyrosine-binding pocket, and this mutant is compromised in its ability to transform NIH 3T3 and pre-B cells, especially at 39.5 degrees C. Our data reveal that loss of p53 or Ink4a/Arf locus products complements the transforming defect of the P120/R273K mutant, but it does not completely restore wild-type function. These results indicate that one important transforming function of v-Abl proteins is overcoming the effects of a functional p53 pathway.


Subject(s)
Abelson murine leukemia virus/pathogenicity , Cell Transformation, Viral , Mutation , Oncogene Proteins v-abl/genetics , Signal Transduction , Tumor Suppressor Protein p53/genetics , 3T3 Cells , Animals , Apoptosis , B-Lymphocytes/virology , Cell Line , Cell Line, Transformed , Humans , Mice , Oncogene Proteins v-abl/chemistry , Oncogene Proteins v-abl/metabolism , Temperature , Tumor Suppressor Protein p53/deficiency , src Homology Domains
19.
Proc Natl Acad Sci U S A ; 96(2): 388-93, 1999 Jan 19.
Article in English | MEDLINE | ID: mdl-9892643

ABSTRACT

A convenient in vitro chemical ligation strategy has been developed that allows folded recombinant proteins to be joined together. This strategy permits segmental, selective isotopic labeling of the product. The src homology type 3 and 2 domains (SH3 and SH2) of Abelson protein tyrosine kinase, which constitute the regulatory apparatus of the protein, were individually prepared in reactive forms that can be ligated together under normal protein-folding conditions to form a normal peptide bond at the ligation junction. This strategy was used to prepare NMR sample quantities of the Abelson protein tyrosine kinase-SH(32) domain pair, in which only one of the domains was labeled with 15N. Mass spectrometry and NMR analyses were used to confirm the structure of the ligated protein, which was also shown to have appropriate ligand-binding properties. The ability to prepare recombinant proteins with selectively labeled segments having a single-site mutation, by using a combination of expression of fusion proteins and chemical ligation in vitro, will increase the size limits for protein structural determination in solution with NMR methods. In vitro chemical ligation of expressed protein domains will also provide a combinatorial approach to the synthesis of linked protein domains.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Protein Folding , Recombinant Fusion Proteins/chemistry , Amino Acid Sequence , Cloning, Molecular , Humans , Mass Spectrometry , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Nitrogen Isotopes , Oncogene Proteins v-abl/chemistry , Protein Engineering , src Homology Domains/genetics
20.
J Virol ; 75(4): 1816-23, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11160680

ABSTRACT

Abelson murine leukemia virus (Ab-MLV) encodes the v-Abl protein tyrosine kinase and induces transformation of immortalized fibroblast lines and pre-B cells. Temperature-sensitive mutations affecting the kinase domain of the protein have demonstrated that the kinase activity is absolutely required for transformation. Despite this requirement, mutations affecting other regions of v-Abl modulate transformation activity. The SH2 domain and the highly conserved FLVRES motif within it form a phosphotyrosine-binding pocket that is required for interactions between the kinase and cellular substrates. To understand the impact of SH2 alterations on Ab-MLV-mediated transformation, we studied the Ab-MLV mutant P120/R273K. This mutant encodes a v-Abl protein in which the beta B5 arginine at the base of the phosphotyrosine-binding pocket has been replaced by a lysine. Unexpectedly, infection of NIH 3T3 or pre-B cells with P120/R273K revealed a temperature-dependent transformation phenotype. At 34 degrees C, P120/R273K transformed about 10-fold fewer cells than wild-type virus of equivalent titer; at 39.5 degrees C, 300-fold fewer NIH 3T3 cells were transformed and pre-B cells were refractory to transformation. Temperature-dependent transformation was accompanied by decreased phosphorylation of Shc, a protein that interacts with the v-Abl SH2 and links the protein to Ras, and decreased induction of c-Myc expression. These data suggest that alteration of the FLVRES pocket affects the ability of v-Abl to interact with at least some of its substrates in a temperature-dependent fashion and identify a novel type of temperature-sensitive Abelson virus.


Subject(s)
Abelson murine leukemia virus/pathogenicity , Cell Transformation, Viral , Mutation , src Homology Domains/genetics , 3T3 Cells , Abelson murine leukemia virus/genetics , Abelson murine leukemia virus/physiology , Animals , B-Lymphocytes/virology , Cell Line, Transformed , Genes, Viral , Mice , Oncogene Proteins v-abl/chemistry , Oncogene Proteins v-abl/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Premenopause , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Temperature , ras Proteins/genetics , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL