158 results on '"Proto-Oncogene Proteins c-abl physiology"'
Search Results
2. c-Abl activates RIPK3 signaling in Gaucher disease.
- Author
-
Yañez MJ, Campos F, Marín T, Klein AD, Futerman AH, Alvarez AR, and Zanlungo S
- Subjects
- Animals, Gaucher Disease genetics, Gaucher Disease metabolism, Glucosylceramidase genetics, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Necroptosis, Neurons metabolism, Phosphorylation, Receptor-Interacting Protein Serine-Threonine Kinases genetics, Signal Transduction, Apoptosis, Gaucher Disease pathology, Glucosylceramidase metabolism, Neurons pathology, Proto-Oncogene Proteins c-abl physiology, Receptor-Interacting Protein Serine-Threonine Kinases metabolism
- Abstract
Gaucher disease (GD) is caused by homozygous mutations in the GBA1 gene, which encodes the lysosomal β-glucosidase (GBA) enzyme. GD affects several organs and tissues, including the brain in certain variants of the disease. Heterozygous GBA1 variants are a major genetic risk factor for developing Parkinson's disease. The RIPK3 kinase is relevant in GD and its deficiency improves the neurological and visceral symptoms in a murine GD model. RIPK3 mediates necroptotic-like cell death: it is unknown whether the role of RIPK3 in GD is the direct induction of necroptosis or if it has a more indirect function by mediating necrosis-independent. Also, the mechanisms that activate RIPK3 in GD are currently unknown. In this study, we show that c-Abl tyrosine kinase participates upstream of RIPK3 in GD. We found that the active, phosphorylated form of c-Abl is increased in several GD models, including patient's fibroblasts and GBA null mice. Furthermore, its pharmacological inhibition with the FDA-approved drug Imatinib decreased RIPK3 signaling. We found that c-Abl interacts with RIPK3, that RIPK3 is phosphorylated at a tyrosine site, and that this phosphorylation is reduced when c-Abl is inhibited. Genetic ablation of c-Abl in neuronal GD and GD mice models significantly reduced RIPK3 activation and MLKL downstream signaling. These results showed that c-Abl signaling is a new upstream pathway that activates RIPK3 and that its inhibition is an attractive therapeutic approach for the treatment of GD., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
3. Targeting ABL1 or ARG Tyrosine Kinases to Restrict HIV-1 Infection in Primary CD4+ T-Cells or in Humanized NSG Mice.
- Author
-
McCarthy SDS, Leontyev D, Nicoletti P, Binnington B, Kozlowski HN, Ostrowski M, Cochrane A, Branch DR, and Wong RW
- Subjects
- Animals, CD4-Positive T-Lymphocytes immunology, Dasatinib therapeutic use, Female, HIV Infections immunology, Humans, Male, Mice, Mice, Inbred NOD, Protein-Tyrosine Kinases physiology, Proto-Oncogene Proteins c-abl physiology, RNA, Small Interfering genetics, HIV Infections drug therapy, HIV-1 drug effects, Protein Kinase Inhibitors therapeutic use, Protein-Tyrosine Kinases antagonists & inhibitors, Proto-Oncogene Proteins c-abl antagonists & inhibitors
- Abstract
Background: Previous studies support dasatinib as a potent inhibitor of HIV-1 replication. However, a functional distinction between 2 kinase targets of the drug, ABL1 and ARG, has not been assessed., Setting: We used primary CD4 T-cells, CD8-depleted peripheral blood mononuclear cells (PBMCs) from a treatment naïve HIV-1 patient, and a humanized mouse model of HIV-1 infection. We assessed the roles of ABL1 and ARG during HIV-1 infection and use of dasatinib as a potential antiviral against HIV-1 in humanized mice., Methods: Primary CD4 T-cells were administered siRNA targeting ABL1 or ARG, then infected with HIV-1 containing luciferase reporter viruses. Quantitative polymerase chain reaction of viral integration of 4 HIV-1 strains was also assessed. CD8-depleted PBMCs were treated for 3 weeks with dasatinib. NSG mice were engrafted with CD34 pluripotent stem cells from human fetal cord blood, and infected with Ba-L virus after 19 weeks. Mice were treated daily with dasatinib starting 5 weeks after infection., Results: siRNA knockdown of ABL1 or ARG had no effect on viral reverse transcripts, but increased 2-LTR circles 2- to 4-fold and reduced viral integration 2- to 12-fold. siRNA knockdown of ARG increased SAMHD1 activation, whereas knockdown of either kinase reduced RNA polymerase II activation. Treating CD8-depleted PBMCs from a treatment-naïve patient with 50 nM of dasatinib for 3 weeks reduced p24 levels by 99.8%. Ba-L (R5)-infected mice injected daily with dasatinib showed a 95.1% reduction in plasma viral load after 2 weeks of treatment., Conclusions: We demonstrate a novel nuclear role for ABL1 and ARG in ex vivo infection experiments, and proof-of-principle use of dasatinib in a humanized mouse model of HIV-1 infection.
- Published
- 2019
- Full Text
- View/download PDF
4. C-Abl Inhibition; A Novel Therapeutic Target for Parkinson's Disease.
- Author
-
Abushouk AI, Negida A, Elshenawy RA, Zein H, Hammad AM, Menshawy A, and Mohamed WMY
- Subjects
- Animals, Humans, Neuroprotection drug effects, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Molecular Targeted Therapy methods, Neuroprotection physiology, Parkinson Disease drug therapy, Parkinson Disease physiopathology, Proto-Oncogene Proteins c-abl antagonists & inhibitors, Proto-Oncogene Proteins c-abl physiology
- Abstract
Parkinson's disease (PD) is the most prevalent movement disorder in the world. The major pathological hallmarks of PD are death of dopaminergic neurons and the formation of Lewy bodies. At the moment, there is no cure for PD; current treatments are symptomatic. Investigators are searching for neuroprotective agents and disease modifying strategies to slow the progress of neurodegeneration. However, due to lack of data about the main pathological sequence of PD, many drug targets failed to provide neuroprotective effects in human trials. Recent evidence suggests the involvement of C-Abelson (c-Abl) tyrosine kinase enzyme in the pathogenesis of PD. Through parkin inactivation, alpha synuclein aggregation, and impaired autophagy of toxic elements. Experimental studies showed that (1) c-Abl activation is involved in neurodegeneration and (2) c-Abl inhibition shows neuroprotective effects and prevents dopaminergic neuronal' death. Current evidence from experimental studies and the first in-human trial shows that c-Abl inhibition holds the promise for neuroprotection against PD and therefore, justifies the movement towards larger clinical trials. In this review article, we discussed the role of c-Abl in PD pathogenesis and the findings of preclinical experiments and the first in-human trial. In addition, based on lessons from the last decade and current preclinical evidence, we provide recommendations for future research in this area., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.org.)
- Published
- 2018
- Full Text
- View/download PDF
5. VEGF165-induced vascular permeability requires NRP1 for ABL-mediated SRC family kinase activation.
- Author
-
Fantin A, Lampropoulou A, Senatore V, Brash JT, Prahst C, Lange CA, Liyanage SE, Raimondi C, Bainbridge JW, Augustin HG, and Ruhrberg C
- Subjects
- Adaptor Proteins, Signal Transducing physiology, Animals, Enzyme Activation, Mice, Mice, Inbred C57BL, Semaphorin-3A physiology, Vascular Endothelial Growth Factor Receptor-2 physiology, Capillary Permeability, Neuropilin-1 physiology, Proto-Oncogene Proteins c-abl physiology, Vascular Endothelial Growth Factor A physiology, src-Family Kinases metabolism
- Abstract
The vascular endothelial growth factor (VEGF) isoform VEGF165 stimulates vascular growth and hyperpermeability. Whereas blood vessel growth is essential to sustain organ health, chronic hyperpermeability causes damaging tissue edema. By combining in vivo and tissue culture models, we show here that VEGF165-induced vascular leakage requires both VEGFR2 and NRP1, including the VEGF164-binding site of NRP1 and the NRP1 cytoplasmic domain (NCD), but not the known NCD interactor GIPC1. In the VEGF165-bound receptor complex, the NCD promotes ABL kinase activation, which in turn is required to activate VEGFR2-recruited SRC family kinases (SFKs). These results elucidate the receptor complex and signaling hierarchy of downstream kinases that transduce the permeability response to VEGF165. In a mouse model with choroidal neovascularisation akin to age-related macular degeneration, NCD loss attenuated vessel leakage without affecting neovascularisation. These findings raise the possibility that targeting NRP1 or its NCD interactors may be a useful therapeutic strategy in neovascular disease to reduce VEGF165-induced edema without compromising vessel growth., (© 2017 Fantin et al.)
- Published
- 2017
- Full Text
- View/download PDF
6. Normal ABL1 is a tumor suppressor and therapeutic target in human and mouse leukemias expressing oncogenic ABL1 kinases.
- Author
-
Dasgupta Y, Koptyra M, Hoser G, Kantekure K, Roy D, Gornicka B, Nieborowska-Skorska M, Bolton-Gillespie E, Cerny-Reiterer S, Müschen M, Valent P, Wasik MA, Richardson C, Hantschel O, van der Kuip H, Stoklosa T, and Skorski T
- Subjects
- Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Apoptosis drug effects, Blast Crisis drug therapy, Blast Crisis enzymology, Blast Crisis pathology, Cell Division drug effects, Cell Line, Tumor, Cytostatic Agents pharmacology, Gene Expression Regulation, Leukemic drug effects, Genomic Instability, Humans, Imatinib Mesylate pharmacology, Imatinib Mesylate therapeutic use, Imidazoles pharmacology, Imidazoles therapeutic use, Leukemia, Experimental drug therapy, Leukemia, Experimental enzymology, Leukemia, Experimental pathology, Leukemia, Myeloid, Chronic-Phase drug therapy, Leukemia, Myeloid, Chronic-Phase enzymology, Leukemia, Myeloid, Chronic-Phase pathology, Mice, Mice, Inbred NOD, Mice, SCID, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins genetics, Neoplasm Proteins physiology, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells enzymology, Oncogene Proteins v-abl antagonists & inhibitors, Oncogene Proteins v-abl genetics, Oncogene Proteins, Fusion antagonists & inhibitors, Oncogene Proteins, Fusion genetics, Oxidative Stress, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins c-abl genetics, Pyridazines pharmacology, Pyridazines therapeutic use, Tumor Suppressor Proteins antagonists & inhibitors, Tumor Suppressor Proteins genetics, Blast Crisis genetics, Genes, Tumor Suppressor, Genes, abl, Leukemia, Experimental genetics, Leukemia, Myeloid, Chronic-Phase genetics, Oncogene Proteins v-abl physiology, Oncogene Proteins, Fusion physiology, Proto-Oncogene Proteins c-abl physiology, Tumor Suppressor Proteins physiology
- Abstract
Leukemias expressing constitutively activated mutants of ABL1 tyrosine kinase (BCR-ABL1, TEL-ABL1, NUP214-ABL1) usually contain at least 1 normal ABL1 allele. Because oncogenic and normal ABL1 kinases may exert opposite effects on cell behavior, we examined the role of normal ABL1 in leukemias induced by oncogenic ABL1 kinases. BCR-ABL1-Abl1(-/-) cells generated highly aggressive chronic myeloid leukemia (CML)-blast phase-like disease in mice compared with less malignant CML-chronic phase-like disease from BCR-ABL1-Abl1(+/+) cells. Additionally, loss of ABL1 stimulated proliferation and expansion of BCR-ABL1 murine leukemia stem cells, arrested myeloid differentiation, inhibited genotoxic stress-induced apoptosis, and facilitated accumulation of chromosomal aberrations. Conversely, allosteric stimulation of ABL1 kinase activity enhanced the antileukemia effect of ABL1 tyrosine kinase inhibitors (imatinib and ponatinib) in human and murine leukemias expressing BCR-ABL1, TEL-ABL1, and NUP214-ABL1. Therefore, we postulate that normal ABL1 kinase behaves like a tumor suppressor and therapeutic target in leukemias expressing oncogenic forms of the kinase., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
7. Are Molecules Involved in Neuritogenesis and Axon Guidance Related to Autism Pathogenesis?
- Author
-
Bakos J, Bacova Z, Grant SG, Castejon AM, and Ostatnikova D
- Subjects
- Autism Spectrum Disorder genetics, Autism Spectrum Disorder pathology, Autism Spectrum Disorder physiopathology, Axonal Transport, Axons physiology, Biomarkers, Brain pathology, Connectome, GTP Phosphohydrolases physiology, Gene Expression Profiling, Growth Cones physiology, Humans, Microtubules physiology, Models, Neurological, Nerve Tissue Proteins physiology, Neuronal Plasticity, Proto-Oncogene Proteins c-abl physiology, RNA, Messenger biosynthesis, RNA, Messenger genetics, Signal Transduction, cdc42 GTP-Binding Protein physiology, Autism Spectrum Disorder etiology, Neurites pathology, Neurogenesis
- Abstract
Autism spectrum disorder is a heterogeneous disease, and numerous alterations of gene expression come into play to attempt to explain potential molecular and pathophysiological causes. Abnormalities of brain development and connectivity associated with alterations in cytoskeletal rearrangement, neuritogenesis and elongation of axons and dendrites might represent or contribute to the structural basis of autism pathology. Slit/Robo signaling regulates cytoskeletal remodeling related to axonal and dendritic branching. Components of its signaling pathway (ABL and Cdc42) are suspected to be molecular bases of alterations of normal development. The present review describes the most important mechanisms underlying neuritogenesis, axon pathfinding and the role of GTPases in neurite outgrowth, with special emphasis on alterations associated with autism spectrum disorders. On the basis of analysis of publicly available microarray data, potential biomarkers of autism are discussed.
- Published
- 2015
- Full Text
- View/download PDF
8. The proto-oncogene c-Src and its downstream signaling pathways are inhibited by the metastasis suppressor, NDRG1.
- Author
-
Liu W, Yue F, Zheng M, Merlot A, Bae DH, Huang M, Lane D, Jansson P, Lui GY, Richardson V, Sahni S, Kalinowski D, Kovacevic Z, and Richardson DR
- Subjects
- Adenocarcinoma pathology, Cell Line, Tumor, Cell Movement, Colonic Neoplasms pathology, Crk-Associated Substrate Protein physiology, Down-Regulation, Enzyme Activation physiology, ErbB Receptors biosynthesis, ErbB Receptors genetics, Gene Expression Regulation, Neoplastic physiology, Genes, src, Humans, Indoles, Male, Phosphorylation, Prostatic Neoplasms pathology, Protein Processing, Post-Translational, Proto-Oncogene Mas, Proto-Oncogene Proteins c-abl physiology, Proto-Oncogene Proteins c-crk physiology, Proto-Oncogene Proteins pp60(c-src) physiology, RNA Interference, RNA, Small Interfering genetics, Recombinant Proteins metabolism, Sulfonamides, p21-Activated Kinases physiology, rac1 GTP-Binding Protein physiology, Cell Cycle Proteins physiology, Intracellular Signaling Peptides and Proteins physiology, Neoplasm Proteins physiology, Proto-Oncogene Proteins pp60(c-src) antagonists & inhibitors, Signal Transduction physiology
- Abstract
N-myc downstream regulated gene-1 (NDRG1) is a potent metastasis suppressor that plays a key role in regulating signaling pathways involved in mediating cancer cell invasion and migration, including those derived from prostate, colon, etc. However, the mechanisms and molecular targets through which NDRG1 reduces cancer cell invasion and migration, leading to inhibition of cancer metastasis, are not fully elucidated. In this investigation, using NDRG1 over-expression models in three tumor cell-types (namely, DU145, PC3MM and HT29) and also NDRG1 silencing in DU145 and HT29 cells, we reveal that NDRG1 decreases phosphorylation of a key proto-oncogene, cellular Src (c-Src), at a well-characterized activating site (Tyr416). NDRG1-mediated down-regulation of EGFR expression and activation were responsible for the decreased phosphorylation of c-Src (Tyr416). Indeed, NDRG1 prevented recruitment of c-Src to EGFR and c-Src activation. Moreover, NDRG1 suppressed Rac1 activity by modulating phosphorylation of a c-Src downstream effector, p130Cas, and its association with CrkII, which acts as a "molecular switch" to activate Rac1. NDRG1 also affected another signaling molecule involved in modulating Rac1 signaling, c-Abl, which then inhibited CrkII phosphorylation. Silencing NDRG1 increased cell migration relative to the control and inhibition of c-Src signaling using siRNA, or a pharmacological inhibitor (SU6656), prevented this increase. Hence, the role of NDRG1 in decreasing cell migration is, in part, due to its inhibition of c-Src activation. In addition, novel pharmacological agents, which induce NDRG1 expression and are currently under development as anti-metastatic agents, markedly increase NDRG1 and decrease c-Src activation. This study leads to important insights into the mechanism involved in inhibiting metastasis by NDRG1 and how to target these pathways with novel therapeutics.
- Published
- 2015
- Full Text
- View/download PDF
9. Imatinib mesylate stimulates low-density lipoprotein receptor-related protein 1-mediated ERK phosphorylation in insulin-producing cells.
- Author
-
Fred RG, Boddeti SK, Lundberg M, and Welsh N
- Subjects
- Animals, Apoptosis drug effects, Cells, Cultured, Gene Knockdown Techniques, Imatinib Mesylate, Insulin-Secreting Cells metabolism, Insulin-Secreting Cells physiology, Low Density Lipoprotein Receptor-Related Protein-1 physiology, MAP Kinase Signaling System physiology, Mitogen-Activated Protein Kinase Kinases drug effects, Mitogen-Activated Protein Kinase Kinases metabolism, Mitogen-Activated Protein Kinase Kinases physiology, Phosphorylation drug effects, Proto-Oncogene Proteins c-abl drug effects, Proto-Oncogene Proteins c-abl physiology, Rats, Benzamides pharmacology, Insulin-Secreting Cells drug effects, Low Density Lipoprotein Receptor-Related Protein-1 drug effects, MAP Kinase Signaling System drug effects, Piperazines pharmacology, Protein-Tyrosine Kinases antagonists & inhibitors, Pyrimidines pharmacology
- Abstract
Low-density lipoprotein receptor-related protein 1 (LRP1) is an endocytic and multi-functional type I cell surface membrane protein, which is known to be phosphorylated by the activated platelet-derived growth factor receptor (PDGFR). The tyrosine kinase inhibitor imatinib, which inhibits PDGFR and c-Abl, and which has previously been reported to counteract β-cell death and diabetes, has been suggested to reduce atherosclerosis by inhibiting PDGFR-induced LRP1 phosphorylation. The aim of the present study was to study LRP1 function in β-cells and to what extent imatinib modulates LRP1 activity. LRP1 and c-Abl gene knockdown was performed by RNAi using rat INS-1 832/13 and human EndoC1-βH1 cells. LRP1 was also antagonized by treatment with the antagonist low-density lipoprotein receptor-related protein associated protein 1 (LRPAP1). We have used PDGF-BB, a PDGFR agonist, and apolipoprotein E (ApoE), an LRP1 agonist, to stimulate the activities of PDGFR and LRP1 respectively. Knockdown or inhibition of LRP1 resulted in increased hydrogen peroxide (H2O2)- or cytokine-induced cell death, and glucose-induced insulin release was lowered in LRP1-silenced cells. These results indicate that LRP1 function is necessary for β-cell function and that LRP1 is adversely affected by challenges to β-cell health. PDGF-BB, or the combination of PDGF-BB+ApoE, induced phosphorylation of extracellular-signal-regulated kinase (ERK), Akt and LRP1. LRP1 silencing blocked this event. Imatinib blocked phosphorylation of LRP1 by PDGFR activation but induced phosphorylation of ERK. LRP1 silencing blocked imatinib-induced phosphorylation of ERK. Sunitinib also blocked LRP1 phosphorylation in response to PDGF-BB and induced phosphorylation of ERK, but this latter event was not affected by LRP1 knockdown. siRNA-mediated knockdown of the imatinib target c-Abl resulted in an increased ERK phosphorylation at basal conditions, with no further increase in response to imatinib. Imatinib-induced cell survival of tunicamycin-treated cells was partially mediated by ERK activation. We have concluded that imatinib promotes LRP1-dependent ERK activation, possibly via inhibition of c-Abl, and that this could contribute to the pro-survival effects of imatinib on β-cells.
- Published
- 2015
- Full Text
- View/download PDF
10. c-Abl tyrosine kinase promotes adipocyte differentiation by targeting PPAR-gamma 2.
- Author
-
Keshet R, Bryansker Kraitshtein Z, Shanzer M, Adler J, Reuven N, and Shaul Y
- Subjects
- 3T3-L1 Cells, Adipocytes drug effects, Adipogenesis drug effects, Animals, Benzamides pharmacology, HEK293 Cells, Humans, Imatinib Mesylate, Mice, Mutation, Missense, NIH 3T3 Cells, PPAR gamma chemistry, PPAR gamma genetics, Phosphorylation, Phosphotyrosine chemistry, Piperazines pharmacology, Point Mutation, Polymorphism, Single Nucleotide, Proline chemistry, Protein Binding, Protein Interaction Mapping, Protein Isoforms biosynthesis, Protein Isoforms genetics, Protein Kinase Inhibitors pharmacology, Protein Processing, Post-Translational, Protein Stability, Protein Structure, Tertiary, Proto-Oncogene Proteins c-abl antagonists & inhibitors, Proto-Oncogene Proteins c-abl chemistry, Pyrimidines pharmacology, Sequence Homology, Amino Acid, Species Specificity, Transcription, Genetic, Adipocytes metabolism, Adipogenesis physiology, PPAR gamma physiology, Proto-Oncogene Proteins c-abl physiology
- Abstract
Adipocyte differentiation, or adipogenesis, is a complex and highly regulated process. A recent proteomic analysis has predicted that the nonreceptor tyrosine kinase Abelson murine leukemia viral oncogene (c-Abl) is a putative key regulator of adipogenesis, but the underlying mechanism remained obscure. We found that c-Abl was activated during the early phase of mouse 3T3-L1 preadipocyte differentiation. Moreover, c-Abl activity was essential and its inhibition blocked differentiation to mature adipocytes. c-Abl directly controlled the expression and activity of the master adipogenic regulator peroxisome proliferator-activator receptor gamma 2 (PPARγ2). PPARγ2 physically associated with c-Abl and underwent phosphorylation on two tyrosine residues within its regulatory activation function 1 (AF1) domain. We demonstrated that this process positively regulates PPARγ2 stability and adipogenesis. Remarkably, c-Abl binding to PPARγ2 required the Pro12 residue that has a phenotypically well-studied common human genetic proline 12 alanine substitution (Pro12Ala) polymorphism. Our findings establish a critical role for c-Abl in adipocyte differentiation and explain the behavior of the known Pro12Ala polymorphism.
- Published
- 2014
- Full Text
- View/download PDF
11. c-Abl stabilizes HDAC2 levels by tyrosine phosphorylation repressing neuronal gene expression in Alzheimer's disease.
- Author
-
Gonzalez-Zuñiga M, Contreras PS, Estrada LD, Chamorro D, Villagra A, Zanlungo S, Seto E, and Alvarez AR
- Subjects
- Epigenesis, Genetic, Gene Expression Regulation, HeLa Cells, Humans, Phosphorylation, Proto-Oncogene Proteins c-abl genetics, Proto-Oncogene Proteins c-abl metabolism, Tyrosine metabolism, Alzheimer Disease genetics, Histone Deacetylase 2 metabolism, Neurons metabolism, Proto-Oncogene Proteins c-abl physiology
- Abstract
In Alzheimer's disease (AD), there is a decrease in neuronal gene expression induced by HDAC2 increase; however, the mechanisms involved are not fully elucidated. Here, we described how the tyrosine kinase c-Abl increases HDAC2 levels, inducing transcriptional repression of synaptic genes. Our data demonstrate that (1) in neurons, c-Abl inhibition with Imatinib prevents the AβO-induced increase in HDAC2 levels; (2) c-Abl knockdown cells show a decrease in HDAC2 levels, while c-Abl overexpression increases them; (3) c-Abl inhibition reduces HDAC2-dependent repression activity and HDAC2 recruitment to the promoter of several synaptic genes, increasing their expression; (4) c-Abl induces tyrosine phosphorylation of HDAC2, a posttranslational modification, affecting both its stability and repression activity; and (5) treatment with Imatinib decreases HDAC2 levels in a transgenic mice model of AD. Our results support the participation of the c-Abl/HDAC2 signaling pathway in the epigenetic blockade of gene expression in AD pathology., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
12. c-Abl activates janus kinase 2 in normal hematopoietic cells.
- Author
-
Tao W, Leng X, Chakraborty SN, Ma H, and Arlinghaus RB
- Subjects
- Animals, Base Sequence, Bone Marrow Cells drug effects, Bone Marrow Cells metabolism, Cell Line, Cell Survival, DNA Primers, Enzyme Activation, Interleukin-3 pharmacology, Mice, Polymerase Chain Reaction, Bone Marrow Cells enzymology, Janus Kinase 2 metabolism, Proto-Oncogene Proteins c-abl physiology
- Abstract
Jak2 is involved in cytokine growth factor-stimulated signal transduction, but the mechanism of its activation is largely unknown. Here, we investigated Jak2 activation in a normal hematopoietic cell line, 32D mouse myeloid cells. The bimolecular fluorescence complementation studies showed that c-Abl formed a stable complex with Jak2 in live cells. Co-immunoprecipitation results showed that c-Abl bound to the βc chain of IL-3/IL-5/GM-CSF receptors. The kinase activities of both c-Abl and Jak2 were stimulated by IL-3 in 32D cells. Decreasing c-Abl protein expression in 32D cells by inducible shRNA decreased Jak2 activity and resulted in the failure of Jak2 activation in response to IL-3. Treatment of IL-3 and serum-starved 32D cells with 1 μM imatinib mysylate inhibited IL-3 stimulated kinase activities of both c-Abl and Jak2. In addition, the kinase-deficient Bcr-Abl mutant (p210K1172R) was defective for activation of Jak2 in 32D cells and impaired IL-3 independent growth, which was rescued by overexpression of c-Abl (+Abl). IL-3 efficiently inhibited apoptosis of 32Dp210K/R+Abl cells induced by imatinib mysylate but not Jak2 kinase inhibitor TG101209. In summary, our findings provide evidence that the kinase function of c-Abl and its C-terminal CT4 region is crucial for its interaction with Jak2 and its activation. c-Abl kinase activity induced by IL-3 is required for IL-3-stimulated Jak2 and Jak1 activation. Our findings reveal a novel regulatory role of c-Abl in Jak2 activation induced by IL-3 cytokine growth factor in 32D hematopoietic cells., (© 2014 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
13. Oxidative stress-induced signaling pathways implicated in the pathogenesis of Parkinson's disease.
- Author
-
Gaki GS and Papavassiliou AG
- Subjects
- Animals, Apoptosis, Basal Ganglia metabolism, Basal Ganglia pathology, Calcium Signaling, Dopaminergic Neurons drug effects, Dopaminergic Neurons metabolism, Drosophila Proteins physiology, Drosophila melanogaster, Humans, Metalloproteases genetics, Metalloproteases physiology, Metals, Heavy adverse effects, Microglia physiology, Mitochondria metabolism, Mitochondrial Proteins genetics, Mitochondrial Proteins physiology, Models, Neurological, Neuroprotective Agents therapeutic use, Neurotoxins toxicity, Parkinson Disease genetics, Parkinson Disease metabolism, Parkinsonian Disorders genetics, Parkinsonian Disorders metabolism, Protein Kinases genetics, Protein Kinases physiology, Proto-Oncogene Proteins c-abl physiology, Receptors, Glutamate physiology, Signal Transduction physiology, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases physiology, Xanthophylls therapeutic use, Oxidative Stress, Parkinson Disease etiology
- Abstract
Parkinson's disease is the second most common neurodegenerative movement disorder; however, its etiology remains elusive. Nevertheless, in vivo observations have concluded that oxidative stress is one of the most common causes in the pathogenesis of Parkinson's disease. It is known that mitochondria play a crucial role in reactive oxygen species-mediated pathways, and several gene products that associate with mitochondrial function are the subject of Parkinson's disease research. The PTEN-induced kinase 1 (PINK1) protects cells from mitochondrial dysfunction and is linked to the autosomal recessive familial form of the disease. PINK1 is a key player in many signaling pathways engaged in mitophagy, apoptosis, or microglial inflammatory response and is induced by oxidative stress. Several proteins participate in mitochondrial networks, and they are associated with PINK1. The E3 ubiquitin ligase Parkin, the protease presenilin-associated rhomboid-like serine protease, the tyrosine kinase c-Abl, the protein kinase MARK2, the protease HtrA2, and the tumor necrosis factor receptor-associated protein 1 (TRAP1) provide different steps of control in protection against oxidative stress. Furthermore, environmental toxins, such as 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, have been identified as contributors to parkinsonism by increasing oxidative stress in dopaminergic neurons. The present review discusses the mechanisms and effects of oxidative stress, the emerging concept of the impact of environmental toxins, and a possible neuroprotective role of the antioxidant astaxanthin in various neurodegenerative disorders with particular emphasis in Parkinson's disease.
- Published
- 2014
- Full Text
- View/download PDF
14. C-Abl inhibitor imatinib enhances insulin production by β cells: c-Abl negatively regulates insulin production via interfering with the expression of NKx2.2 and GLUT-2.
- Author
-
Xia CQ, Zhang P, Li S, Yuan L, Xia T, Xie C, and Clare-Salzler MJ
- Subjects
- Animals, Cell Line, Gene Expression Regulation, Glucose metabolism, Glucose Transporter Type 2 genetics, Homeobox Protein Nkx-2.2, Homeodomain Proteins genetics, Imatinib Mesylate, Insulin genetics, Leukemia, Myeloid metabolism, Mice, Peptide Biosynthesis drug effects, Proto-Oncogene Proteins c-abl genetics, Proto-Oncogene Proteins c-abl physiology, RNA, Small Interfering metabolism, Trans-Activators metabolism, Transcription Factors genetics, Zebrafish Proteins, Benzamides pharmacology, Glucose Transporter Type 2 metabolism, Homeodomain Proteins metabolism, Insulin biosynthesis, Insulin-Secreting Cells drug effects, Piperazines pharmacology, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-abl antagonists & inhibitors, Pyrimidines pharmacology, Transcription Factors metabolism
- Abstract
Chronic myelogenous leukemia patients treated with tyrosine kinase inhibitor, Imatinib, were shown to have increased serum levels of C-peptide. Imatinib specifically inhibits the tyrosine kinase, c-Abl. However, the mechanism of how Imatinib treatment can lead to increased insulin level is unclear. Specifically, there is little investigation into whether Imatinib directly affects β cells to promote insulin production. In this study, we showed that Imatinib significantly induced insulin expression in both glucose-stimulated and resting β cells. In line with this finding, c-Abl knockdown by siRNA and overexpression of c-Abl markedly enhanced and inhibited insulin expression in β cells, respectively. Unexpectedly, high concentrations of glucose significantly induced c-Abl expression, suggesting c-Abl may play a role in balancing insulin production during glucose stimulation. Further studies demonstrated that c-Abl inhibition did not affect the major insulin gene transcription factor, pancreatic and duodenal homeobox-1 (PDX-1) expression. Of interest, inhibition of c-Abl enhanced NKx2.2 and overexpression of c-Abl in β cells markedly down-regulated NKx2.2, which is a positive regulator for insulin gene expression. Additionally, we found that c-Abl inhibition significantly enhanced the expression of glucose transporter GLUT2 on β cells. Our study demonstrates a previously unrecognized mechanism that controls insulin expression through c-Abl-regulated NKx2.2 and GLUT2. Therapeutic targeting β cell c-Abl could be employed in the treatment of diabetes or β cell tumor, insulinoma.
- Published
- 2014
- Full Text
- View/download PDF
15. The capable ABL: what is its biological function?
- Author
-
Wang JY
- Subjects
- Animals, Humans, Mice, Models, Biological, Mutation, Nuclear Localization Signals genetics, Oncogene Proteins v-abl antagonists & inhibitors, Oncogene Proteins v-abl genetics, Protein Kinase Inhibitors pharmacology, Protein-Tyrosine Kinases antagonists & inhibitors, Proto-Oncogene Mas, Proto-Oncogene Proteins c-abl deficiency, Proto-Oncogene Proteins c-abl genetics, Signal Transduction, Genes, abl, Oncogene Proteins v-abl physiology, Proto-Oncogene Proteins c-abl physiology
- Abstract
The mammalian ABL1 gene encodes the ubiquitously expressed nonreceptor tyrosine kinase ABL. In response to growth factors, cytokines, cell adhesion, DNA damage, oxidative stress, and other signals, ABL is activated to stimulate cell proliferation or differentiation, survival or death, retraction, or migration. ABL also regulates specialized functions such as antigen receptor signaling in lymphocytes, synapse formation in neurons, and bacterial adhesion to intestinal epithelial cells. Although discovered as the proto-oncogene from which the Abelson leukemia virus derived its Gag-v-Abl oncogene, recent results have linked ABL kinase activation to neuronal degeneration. This body of knowledge on ABL seems confusing because it does not fit the one-gene-one-function paradigm. Without question, ABL capabilities are encoded by its gene sequence and that molecular blueprint designs this kinase to be regulated by subcellular location-dependent interactions with inhibitors and substrate activators. Furthermore, ABL shuttles between the nucleus and the cytoplasm where it binds DNA and actin--two biopolymers with fundamental roles in almost all biological processes. Taken together, the cumulated results from analyses of ABL structure-function, ABL mutant mouse phenotypes, and ABL substrates suggest that this tyrosine kinase does not have its own agenda but that, instead, it has evolved to serve a variety of tissue-specific and context-dependent biological functions.
- Published
- 2014
- Full Text
- View/download PDF
16. A humanin analog decreases oxidative stress and preserves mitochondrial integrity in cardiac myoblasts.
- Author
-
Klein LE, Cui L, Gong Z, Su K, and Muzumdar R
- Subjects
- Animals, Antioxidants pharmacology, Catalase metabolism, Gene Knockdown Techniques, Glutathione Peroxidase metabolism, Hydrogen Peroxide pharmacology, Membrane Potential, Mitochondrial drug effects, Mitochondria drug effects, Mitochondria metabolism, Myoblasts, Cardiac physiology, Neuroprotective Agents pharmacology, Protein-Tyrosine Kinases physiology, Proto-Oncogene Proteins c-abl physiology, Rats, Reactive Oxygen Species metabolism, Intracellular Signaling Peptides and Proteins pharmacology, Myoblasts, Cardiac drug effects, Myocardial Reperfusion Injury prevention & control, Oxidative Stress drug effects, Peptides pharmacology
- Abstract
A potent analog (HNG) of the endogenous peptide humanin protects against myocardial ischemia-reperfusion (MI-R) injury in vivo, decreasing infarct size and improving cardiac function. Since oxidative stress contributes to the damage from MI-R we tested the hypotheses that: (1) HNG offers cardioprotection through activation of antioxidant defense mechanisms leading to preservation of mitochondrial structure and that, (2) the activity of either of a pair of non-receptor tyrosine kinases, c-Abl and Arg is required for this protection. Rat cardiac myoblasts (H9C2 cells) were exposed to nanomolar concentrations of HNG and to hydrogen peroxide (H2O2). Cells treated with HNG in the presence of H2O2 demonstrated reduced intracellular reactive oxygen species (ROS), preserved mitochondrial membrane potential, ATP levels and mitochondrial structure. HNG induced activation of catalase and glutathione peroxidase (GPx) within 5 min and decreased the ratio of oxidized to reduced glutathione within 30 min. siRNA knockdown of both Abl and Arg, but neither alone, abolished the HNG-mediated reduction of ROS in myoblasts exposed to H2O2. These findings demonstrate an HNG-mediated, Abl- and Arg-dependent, rapid and sustained activation of critical cellular defense systems and attenuation of oxidative stress, providing mechanistic insights into the observed HNG-mediated cardioprotection in vivo., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
17. Role of Abl in airway hyperresponsiveness and airway remodeling.
- Author
-
Cleary RA, Wang R, Wang T, and Tang DD
- Subjects
- Animals, Asthma chemically induced, Asthma metabolism, Benzamides pharmacology, Bronchi drug effects, Bronchi metabolism, Bronchi pathology, Bronchial Hyperreactivity chemically induced, Bronchial Hyperreactivity metabolism, Cells, Cultured, Chemokine CCL2 metabolism, Disease Models, Animal, Female, Humans, Imatinib Mesylate, In Vitro Techniques, Interleukin-13 metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Myocytes, Smooth Muscle pathology, Ovalbumin adverse effects, Piperazines pharmacology, Proliferating Cell Nuclear Antigen metabolism, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-abl deficiency, Proto-Oncogene Proteins c-abl genetics, Pyrimidines pharmacology, Airway Remodeling physiology, Asthma physiopathology, Bronchial Hyperreactivity physiopathology, Myocytes, Smooth Muscle physiology, Proto-Oncogene Proteins c-abl physiology
- Abstract
Background: Asthma is a chronic disease that is characterized by airway hyperresponsiveness and airway remodeling. The underlying mechanisms that mediate the pathological processes are not fully understood. Abl is a non-receptor protein tyrosine kinase that has a role in the regulation of smooth muscle contraction and smooth muscle cell proliferation in vitro. The role of Abl in airway hyperresponsiveness and airway remodeling in vivo is largely unknown., Methods: To evaluate the role of Abl in asthma pathology, we assessed the expression of Abl in airway tissues from the ovalbumin sensitized and challenged mouse model, and human asthmatic airway smooth muscle cells. In addition, we generated conditional knockout mice in which Abl expression in smooth muscle was disrupted, and then evaluated the effects of Abl conditional knockout on airway resistance, smooth muscle mass, cell proliferation, IL-13 and CCL2 in the mouse model of asthma. Furthermore, we determined the effects of the Abl pharmacological inhibitors imatinib and GNF-5 on these processes in the animal model of asthma., Results: The expression of Abl was upregulated in airway tissues of the animal model of asthma and in airway smooth muscle cells of patients with severe asthma. Conditional knockout of Abl attenuated airway resistance, smooth muscle mass and staining of proliferating cell nuclear antigen in the airway of mice sensitized and challenged with ovalbumin. Interestingly, conditional knockout of Abl did not affect the levels of IL-13 and CCL2 in bronchoalveolar lavage fluid of animals treated with ovalbumin. However, treatment with imatinib and GNF-5 inhibited the ovalbumin-induced increase in IL-13 and CCL2 as well as airway resistance and smooth muscle growth in animals., Conclusions: These results suggest that the altered expression of Abl in airway smooth muscle may play a critical role in the development of airway hyperresponsiveness and airway remodeling in asthma. Our findings support the concept that Abl may be a novel target for the development of new therapy to treat asthma.
- Published
- 2013
- Full Text
- View/download PDF
18. Role of ABL family kinases in cancer: from leukaemia to solid tumours.
- Author
-
Greuber EK, Smith-Pearson P, Wang J, and Pendergast AM
- Subjects
- Carcinoma enzymology, Humans, Leukemia enzymology, Protein Kinase Inhibitors therapeutic use, Signal Transduction, Carcinoma etiology, Carcinoma therapy, Leukemia etiology, Leukemia therapy, Proto-Oncogene Proteins c-abl physiology
- Abstract
The Abelson (ABL) family of nonreceptor tyrosine kinases, ABL1 and ABL2, transduces diverse extracellular signals to protein networks that control proliferation, survival, migration and invasion. ABL1 was first identified as an oncogene required for the development of leukaemias initiated by retroviruses or chromosome translocations. The demonstration that small-molecule ABL kinase inhibitors could effectively treat chronic myeloid leukaemia opened the door to the era of targeted cancer therapies. Recent reports have uncovered roles for ABL kinases in solid tumours. Enhanced ABL expression and activation in some solid tumours, together with altered cell polarity, invasion or growth induced by activated ABL kinases, suggest that drugs targeting these kinases may be useful for treating selected solid tumours.
- Published
- 2013
- Full Text
- View/download PDF
19. Genetic disruption of Abl nuclear import reduces renal apoptosis in a mouse model of cisplatin-induced nephrotoxicity.
- Author
-
Sridevi P, Nhiayi MK, and Wang JY
- Subjects
- Acute Kidney Injury chemically induced, Acute Kidney Injury physiopathology, Animals, Apoptosis physiology, Apoptosis Regulatory Proteins physiology, Benzamides pharmacology, Disease Models, Animal, Female, Imatinib Mesylate, Kidney Tubules, Proximal drug effects, Kidney Tubules, Proximal physiopathology, Male, Mice, Mice, Knockout, Mutation genetics, Nuclear Localization Signals deficiency, Nuclear Localization Signals genetics, Nuclear Localization Signals physiology, Piperazines pharmacology, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-abl deficiency, Pyrimidines pharmacology, STAT1 Transcription Factor physiology, Tumor Suppressor Protein p53 deficiency, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 physiology, Tumor Suppressor Proteins physiology, Acute Kidney Injury pathology, Apoptosis genetics, Cisplatin adverse effects, Kidney Tubules, Proximal pathology, Proto-Oncogene Proteins c-abl genetics, Proto-Oncogene Proteins c-abl physiology
- Abstract
DNA damage activates nuclear Abl tyrosine kinase to stimulate intrinsic apoptosis in cancer cell lines and mouse embryonic stem cells. To examine the in vivo function of nuclear Abl in apoptosis, we generated Abl-μNLS (μ, mutated in nuclear localization signals) mice. We show here that cisplatin-induced apoptosis is defective in the renal proximal tubule cells (RPTC) from the Abl(μ/μ) mice. When injected with cisplatin, we found similar levels of platinum in the Abl(+/+) and the Abl(μ/μ) kidneys, as well as similar initial inductions of p53 and PUMAα expression. However, the accumulation of p53 and PUMAα could not be sustained in the Abl(μ/μ) kidneys, leading to reductions in renal apoptosis and tubule damage. Co-treatment of cisplatin with the Abl kinase inhibitor, imatinib, reduced the accumulation of p53 and PUMAα in the Abl(+/+) but not in the Abl(μ/μ) kidneys. The residual apoptosis in the Abl(μ/μ) mice was not further reduced in the Abl(μ/μ); p53(-/-) double-mutant mice, suggesting that nuclear Abl and p53 are epistatic to each other in this apoptosis response. Although apoptosis and tubule damage were reduced, cisplatin-induced increases in phospho-Stat-1 and blood urea nitrogen were similar between the Abl(+/+) and the Abl(μ/μ) kidneys, indicating that RPTC apoptosis is not the only factor in cisplatin-induced nephrotoxicity. These results provide in vivo evidence for the pro-apoptotic function of Abl, and show that its nuclear localization and tyrosine kinase activity are both required for the sustained expression of p53 and PUMAα in cisplatin-induced renal apoptosis.
- Published
- 2013
- Full Text
- View/download PDF
20. Met acts through Abl to regulate p53 transcriptional outcomes and cell survival in the developing liver.
- Author
-
Furlan A, Lamballe F, Stagni V, Hussain A, Richelme S, Prodosmo A, Moumen A, Brun C, Del Barco Barrantes I, Arthur JS, Koleske AJ, Nebreda AR, Barilà D, and Maina F
- Subjects
- Animals, Cell Survival, Cyclin-Dependent Kinase Inhibitor p21 genetics, Mice, Phosphorylation, Proto-Oncogene Proteins c-bcl-2 genetics, Proto-Oncogene Proteins c-mdm2 genetics, p38 Mitogen-Activated Protein Kinases metabolism, Hepatocytes physiology, Liver embryology, Proto-Oncogene Proteins c-abl physiology, Proto-Oncogene Proteins c-met physiology, Transcription, Genetic, Tumor Suppressor Protein p53 physiology
- Abstract
Background & Aims: Genetic studies indicate that distinct signaling modulators are each necessary but not individually sufficient for embryonic hepatocyte survival in vivo. Nevertheless, how signaling players are interconnected into functional circuits and how they coordinate the balance of cell survival and death in developing livers are still major unresolved issues. In the present study, we examined the modulation of the p53 pathway by HGF/Met in embryonic livers., Methods: We combined pharmacological and genetic approaches to biochemically and functionally evaluate p53 pathway modulation in primary embryonic hepatocytes and in developing livers. RT-PCR arrays were applied to investigate the selectivity of p53 transcriptional response triggered by Met., Results: Met recruits p53 to regulate the liver developmental program, by qualitatively modulating its transcriptional properties: turning on the Mdm2 survival gene, while keeping death and cell-cycle arrest genes Pmaip1 and p21 silent. We investigated the mechanism leading to p53 regulation by Met and found that Abl and p38MAPK are required for p53 phosphorylation on S(389), Mdm2 upregulation, and hepatocyte survival. Alteration of this signaling mechanism switches p53 properties, leading to p53-dependent cell death in embryonic livers. RT-PCR array studies affirmed the ability of the Met-Abl-p53 axis to modulate the expression of distinct genes that can be regulated by p53., Conclusions: A signaling circuit involving Abl and p38MAPK is required downstream of Met for the survival of embryonic hepatocytes, via qualitative regulation of the p53 transcriptional response, by switching its proapoptotic into survival properties., (Copyright © 2012 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
21. Abl-1-bridged tyrosine phosphorylation of VASP by Abelson kinase impairs association of VASP to focal adhesions and regulates leukaemic cell adhesion.
- Author
-
Maruoka M, Sato M, Yuan Y, Ichiba M, Fujii R, Ogawa T, Ishida-Kitagawa N, Takeya T, and Watanabe N
- Subjects
- Adaptor Proteins, Signal Transducing chemistry, Adaptor Proteins, Signal Transducing genetics, Animals, CHO Cells, Cell Adhesion genetics, Cell Adhesion Molecules genetics, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Cells, Cultured, Cricetinae, Cricetulus, Cytoskeletal Proteins, Focal Adhesions genetics, Fusion Proteins, bcr-abl genetics, Fusion Proteins, bcr-abl metabolism, Fusion Proteins, bcr-abl physiology, HEK293 Cells, Humans, K562 Cells, Leukemia, Experimental enzymology, Leukemia, Experimental genetics, Mice, Microfilament Proteins genetics, NIH 3T3 Cells, Phosphoproteins genetics, Phosphorylation genetics, Protein Binding genetics, Proto-Oncogene Proteins c-abl genetics, Proto-Oncogene Proteins c-abl physiology, Tyrosine metabolism, Xenopus laevis, Vasodilator-Stimulated Phosphoprotein, Adaptor Proteins, Signal Transducing metabolism, Cell Adhesion Molecules metabolism, Focal Adhesions metabolism, Leukemia, Experimental metabolism, Microfilament Proteins metabolism, Phosphoproteins metabolism, Proto-Oncogene Proteins c-abl metabolism
- Abstract
Mena [mammalian Ena (Enabled)]/VASP (vasodilator-stimulated phosphoprotein) proteins are the homologues of Drosophila Ena. In Drosophila, Ena is a substrate of the tyrosine kinase DAbl (Drosophila Abl). However, the link between Abl and the Mena/VASP family is not fully understood in mammals. We previously reported that Abi-1 (Abl interactor 1) promotes phosphorylation of Mena and BCAP (B-cell adaptor for phosphoinositide 3-kinase) by bridging the interaction between c-Abl and the substrate. In the present study we have identified VASP, another member of the Mena/VASP family, as an Abi-1-bridged substrate of Abl. VASP is phosphorylated by Abl when Abi-1 is co-expressed. We also found that VASP interacted with Abi-1 both in vitro and in vivo. VASP was tyrosine-phosphorylated in Bcr-Abl-positive leukaemic cells in an Abi-1-dependent manner. Co-expression of c-Abl and Abi-1 or the phosphomimetic Y39D mutation in VASP resulted in less accumulation of VASP at focal adhesions. VASP Y39D had a reduced affinity to the proline-rich region of zyxin. Interestingly, overexpression of both phosphomimetic and unphosphorylated forms of VASP, but not wild-type VASP, impaired adhesion of K562 cells to fibronectin. These results suggest that the phosphorylation and dephosphorylation cycle of VASP by the Abi-1-bridged mechanism regulates association of VASP with focal adhesions, which may regulate adhesion of Bcr-Abl-transformed leukaemic cells.
- Published
- 2012
- Full Text
- View/download PDF
22. ABL1 regulates spindle orientation in adherent cells and mammalian skin.
- Author
-
Matsumura S, Hamasaki M, Yamamoto T, Ebisuya M, Sato M, Nishida E, and Toyoshima F
- Subjects
- Animals, Cell Adhesion, Epidermis metabolism, HeLa Cells, Humans, Metaphase, Mice, Mice, Knockout, Phosphorylation, Proto-Oncogene Proteins c-abl genetics, RNA Interference, Signal Transduction, Time Factors, Tyrosine chemistry, Gene Expression Regulation, Proto-Oncogene Proteins c-abl physiology, Skin metabolism, Spindle Apparatus
- Abstract
Despite the growing evidence for the regulated spindle orientation in mammals, a systematic approach for identifying the responsible genes in mammalian cells has not been established. Here we perform a kinase-targeting RNAi screen in HeLa cells and identify ABL1 as a novel regulator of spindle orientation. Knockdown of ABL1 causes the cortical accumulation of Leu-Gly-Asn repeat-enriched-protein (LGN), an evolutionarily conserved regulator of spindle orientation. This results in the LGN-dependent spindle rotation and spindle misorientation. In vivo inactivation of ABL1 by a pharmacological inhibitor or by ablation of the abl1 gene causes spindle misorientation and LGN mislocalization in mouse epidermis. Furthermore, ABL1 directly phosphorylates NuMA, a binding partner of LGN, on tyrosine 1774. This phosphorylation maintains the cortical localization of NuMA during metaphase, and ensures the LGN/NuMA-dependent spindle orientation control. This study provides a novel approach to identify genes regulating spindle orientation in mammals and uncovers new signalling pathways for this mechanism.
- Published
- 2012
- Full Text
- View/download PDF
23. Moderate caveolin-1 downregulation prevents NADPH oxidase-dependent endothelial nitric oxide synthase uncoupling by angiotensin II in endothelial cells.
- Author
-
Lobysheva I, Rath G, Sekkali B, Bouzin C, Feron O, Gallez B, Dessy C, and Balligand JL
- Subjects
- Animals, Cells, Cultured, Down-Regulation, Endothelial Cells drug effects, Hemoglobins metabolism, Humans, Hypertension prevention & control, Male, Mice, Nitric Oxide biosynthesis, Proto-Oncogene Proteins c-abl physiology, Reactive Oxygen Species metabolism, Superoxides metabolism, Angiotensin II pharmacology, Caveolin 1 physiology, Endothelial Cells metabolism, NADPH Oxidases physiology, Nitric Oxide Synthase Type III physiology
- Abstract
Objective: We analyzed the role of caveolin-1 (Cav-1) in the cross-talk between NADPH oxidase and endothelial nitric oxide synthase (eNOS) signaling in endothelial caveolae., Methods and Results: In intact endothelial cells, angiotensin II (AII) concurrently increased NO and O(2)(-·) production (to 158±12% and 209±5% of control). NO production was sensitive to inhibition of NADPH oxidase and small interfering RNA downregulation of nonreceptor tyrosine kinase cAbl. Reciprocally, N-nitro-l-arginine methyl ester, a NOS inhibitor, partly inhibited O(2)(-·) stimulated by AII (by 47±11%), indicating eNOS uncoupling, as confirmed by increased eNOS monomer/dimer ratio (by 35%). In endothelial cell fractions separated by isopycnic ultracentrifugation, AII promoted colocalization of cAbl and the NADPH oxidase subunit p47phox with eNOS to Cav-1-enriched fractions, as confirmed by proximity ligation assay. Downregulation of Cav-1 by small interfering RNA (to 50%), although it preserved eNOS confinement, inhibited AII-stimulated p47phox translocation and NADPH oxidase activity in Cav-1-enriched fractions and reversed eNOS uncoupling. AII infusion produced hypertension and decreased blood hemoglobin-NO in Cav-1(+/+) mice but not in heterozygote Cav-1(+/-) mice with similar Cav-1 reduction., Conclusions: Cav-1 critically regulates reactive oxygen species-dependent eNOS activation but also eNOS uncoupling in response to AII, underlining the possibility to treat endothelial dysfunction by modulating Cav-1 abundance.
- Published
- 2011
- Full Text
- View/download PDF
24. c-Abl-mediated tyrosine phosphorylation of the T-bet DNA-binding domain regulates CD4+ T-cell differentiation and allergic lung inflammation.
- Author
-
Chen A, Lee SM, Gao B, Shannon S, Zhu Z, and Fang D
- Subjects
- Animals, Cell Line, DNA-Binding Proteins metabolism, Humans, Mice, Mice, Knockout, Phosphorylation physiology, Pneumonia, Proto-Oncogene Proteins c-abl physiology, Th2 Cells cytology, Th2 Cells immunology, Transcriptional Activation, Tyrosine metabolism, T-bet Transcription Factor, CD4-Positive T-Lymphocytes cytology, Cell Differentiation, Proto-Oncogene Proteins c-abl metabolism, T-Box Domain Proteins metabolism
- Abstract
The tyrosine kinase c-Abl is required for full activation of T cells, while its role in T-cell differentiation has not been characterized. We report that c-Abl deficiency skews CD4(+) T cells to type 2 helper T cell (Th2) differentiation, and c-Abl(-/-) mice are more susceptible to allergic lung inflammation. c-Abl interacts with and phosphorylates T-bet, a Th1 lineage transcription factor. c-Abl-mediated phosphorylation enhances the transcriptional activation of T-bet. Interestingly, three tyrosine residues within the T-bet DNA-binding domain are the predominant sites of phosphorylation by c-Abl. Mutation of these tyrosine residues inhibits the promoter DNA-binding activity of T-bet. c-Abl regulates Th cell differentiation in a T-bet-dependent manner because genetic deletion of T-bet in CD4(+) T cells abolishes c-Abl-deficiency-mediated enhancement of Th2 differentiation. Reintroduction of T-bet-null CD4(+) T cells with wild-type T-bet, but not its tyrosine mutant, rescues gamma interferon (IFN-γ) production and inhibits Th2 cytokine production. Therefore, c-Abl catalyzes tyrosine phosphorylation of the DNA-binding domain of T-bet to regulate CD4(+) T cell differentiation.
- Published
- 2011
- Full Text
- View/download PDF
25. c-Abl tyrosine kinase modulates tau pathology and Cdk5 phosphorylation in AD transgenic mice.
- Author
-
Cancino GI, Perez de Arce K, Castro PU, Toledo EM, von Bernhardi R, and Alvarez AR
- Subjects
- Alzheimer Disease drug therapy, Amyloid beta-Protein Precursor physiology, Animals, Cells, Cultured, Disease Models, Animal, Hippocampus pathology, Hippocampus physiopathology, Humans, Mice, Mice, Transgenic, Phosphorylation physiology, Proto-Oncogene Proteins c-abl antagonists & inhibitors, Rats, Rats, Sprague-Dawley, tau Proteins antagonists & inhibitors, Alzheimer Disease enzymology, Alzheimer Disease pathology, Hippocampus enzymology, Proto-Oncogene Proteins c-abl physiology, tau Proteins metabolism, tau Proteins toxicity
- Abstract
The c-Abl tyrosine kinase is an important link in signal transduction pathways that promote cytoskeletal rearrangement and apoptotic signalling. We have previously shown that amyloid-β-peptide (Aβ) activates c-Abl. Herein we show that c-Abl participates in Aβ-induced tau phosphorylation through Cdk5 activation. We found that intraperitoneal administration of STI571, a specific inhibitor for c-Abl kinase, decreased tau phosphorylation in the APPswe/PSEN1ΔE9 transgenic mouse brain. In addition, when neurons were treated with Aβ we observed: (i) an increase in active c-Abl and tau phosphorylation, (ii) the prevention of tau phosphorylation by STI571 and (iii) the inhibition of c-Abl expression by shRNA, as well as the expression of a c-Abl kinase death mutant, decreased AT8 and PHF1 signals. Furthermore, the increase of c-Abl was associated with Tyr15 phosphorylation of Cdk5 and its association with c-Abl. Brains from APPswe/PSEN1ΔE9 mice showed higher levels of c-Abl and phospho-Cdk5 than wild-type mice. Moreover, STI571 treatment decreased the phospho-Cdk5 levels. Together, the evidence suggests that activation of c-Abl by Aβ promotes tau phosphorylation through Tyr15 phosphorylation-mediated Cdk5 activation., (Copyright © 2009 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
26. c-Abl regulates Mcl-1 gene expression in chronic lymphocytic leukemia cells.
- Author
-
Allen JC, Talab F, Zuzel M, Lin K, and Slupsky JR
- Subjects
- Benzamides, Humans, Imatinib Mesylate, Myeloid Cell Leukemia Sequence 1 Protein, Piperazines pharmacology, Proto-Oncogene Proteins c-abl antagonists & inhibitors, Proto-Oncogene Proteins c-bcl-2 genetics, Pyrimidines pharmacology, RNA, Messenger analysis, RNA, Messenger drug effects, RNA, Small Interfering pharmacology, STAT3 Transcription Factor analysis, STAT3 Transcription Factor metabolism, Tumor Cells, Cultured, Gene Expression Regulation, Leukemic, Leukemia, Lymphocytic, Chronic, B-Cell genetics, Proto-Oncogene Proteins c-abl physiology, Proto-Oncogene Proteins c-bcl-2 physiology
- Abstract
Chronic lymphocytic leukemia (CLL) is a malignancy characterized by clonal expansion of mature B cells that are resistant to apoptosis. This resistance to apoptosis partly results from Mcl-1 expression because high levels of this protein in CLL cells correlate with poor disease prognosis and resistance to chemotherapy. Thus, understanding the mechanism(s) regulating Mcl-1 expression in CLL cells may be useful in the development of new therapies for this incurable disease. In the present study, we show a strong relationship between c-Abl and Mcl-1 expression in CLL cells. We show that treatment of CLL cells with Abl-specific siRNA or with imatinib, to inhibit c-Abl activity, results in the down-regulation of Mcl-1 protein and mRNA. A major regulator of Mcl-1 gene expression is STAT3. Our data show that CLL cells expressing high levels of c-Abl also show elevated levels of phospho-STAT3, and that STAT3 phosphorylation in CLL cells is dependent on c-Abl activity. However, STAT3 phosphorylation by c-Abl requires activation of nuclear factor-κB, secretion of autocrine interleukin-6, and active protein kinase C. Taken together, our data demonstrate the mechanism involved in c-Abl regulation of Mcl-1 expression in CLL cells, and suggest that c-Abl inhibition has therapeutic application in the treatment of this disease.
- Published
- 2011
- Full Text
- View/download PDF
27. An evolutionarily acquired genotoxic response discriminates MyoD from Myf5, and differentially regulates hypaxial and epaxial myogenesis.
- Author
-
Innocenzi A, Latella L, Messina G, Simonatto M, Marullo F, Berghella L, Poizat C, Shu CW, Wang JY, Puri PL, and Cossu G
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins, Biological Evolution, Cell Cycle Proteins metabolism, Cell Differentiation, Cells, Cultured, Coculture Techniques, Cross-Linking Reagents toxicity, DNA Damage, DNA-Binding Proteins metabolism, Etoposide toxicity, Female, Gene Knockdown Techniques, Methyl Methanesulfonate toxicity, Mice embryology, Mitomycin toxicity, MyoD Protein genetics, Myogenic Regulatory Factor 5 genetics, Phosphorylation, Pregnancy, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-abl physiology, RNA Interference, Somites drug effects, Somites metabolism, Tumor Suppressor Proteins metabolism, Muscle Development drug effects, Mutagens toxicity, MyoD Protein metabolism, Myogenic Regulatory Factor 5 metabolism
- Abstract
Despite having distinct expression patterns and phenotypes in mutant mice, the myogenic regulatory factors Myf5 and MyoD have been considered to be functionally equivalent. Here, we report that these factors have a different response to DNA damage, due to the presence in MyoD and absence in Myf5 of a consensus site for Abl-mediated tyrosine phosphorylation that inhibits MyoD activity in response to DNA damage. Genotoxins failed to repress skeletal myogenesis in MyoD-null embryos; reintroduction of wild-type MyoD, but not mutant Abl phosphorylation-resistant MyoD, restored the DNA-damage-dependent inhibition of muscle differentiation. Conversely, introduction of the Abl-responsive phosphorylation motif converts Myf5 into a DNA-damage-sensitive transcription factor. Gene-dosage-dependent reduction of Abl kinase activity in MyoD-expressing cells attenuated the DNA-damage-dependent inhibition of myogenesis. The presence of a DNA-damage-responsive phosphorylation motif in vertebrate, but not in invertebrate MyoD suggests an evolved response to environmental stress, originated from basic helix-loop-helix gene duplication in vertebrate myogenesis.
- Published
- 2011
- Full Text
- View/download PDF
28. c-Abl tyrosine kinase in the DNA damage response: cell death and more.
- Author
-
Meltser V, Ben-Yehoyada M, and Shaul Y
- Subjects
- Antibiotics, Antineoplastic pharmacology, Ataxia Telangiectasia Mutated Proteins, Cell Cycle Proteins metabolism, DNA Repair, DNA-Binding Proteins metabolism, Doxorubicin pharmacology, Humans, MRE11 Homologue Protein, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-abl physiology, Signal Transduction, Tumor Suppressor Proteins metabolism, Apoptosis, DNA Damage, Proto-Oncogene Proteins c-abl metabolism
- Published
- 2011
- Full Text
- View/download PDF
29. A positive role for c-Abl in Atm and Atr activation in DNA damage response.
- Author
-
Wang X, Zeng L, Wang J, Chau JF, Lai KP, Jia D, Poonepalli A, Hande MP, Liu H, He G, He L, and Li B
- Subjects
- Animals, Antibiotics, Antineoplastic pharmacology, Apoptosis, Ataxia Telangiectasia Mutated Proteins, Cell Line, Checkpoint Kinase 1, Checkpoint Kinase 2, DNA Breaks, Double-Stranded, DNA Repair, Doxorubicin pharmacology, Fibroblasts metabolism, Mice, Mice, Knockout, Nuclear Proteins metabolism, Phosphorylation, Protein Kinases metabolism, Proto-Oncogene Proteins c-abl genetics, Proto-Oncogene Proteins c-abl metabolism, Tumor Protein p73, Tumor Suppressor Protein p53 metabolism, Cell Cycle Proteins metabolism, DNA Damage, DNA-Binding Proteins metabolism, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-abl physiology, Tumor Suppressor Proteins metabolism
- Abstract
DNA damage triggers Atm- and/or Atr-dependent signaling pathways to control cell cycle progression, apoptosis, and DNA repair. However, how Atm and Atr are activated is not fully understood. One of the downstream targets of Atm is non-receptor tyrosine kinase c-Abl, which is phosphorylated and activated by Atm. The current view is that c-Abl relays pro-apoptotic signals from Atm to p73 and p53. Here we show that c-Abl deficiency resulted in a broad spectrum of defects in cell response to genotoxic stress, including activation of Chk1 and Chk2, activation of p53, nuclear foci formation, apoptosis, and DNA repair, suggesting that c-Abl might also act upstream of the DNA damage-activated signaling cascades in addition to its role in p73 and p53 regulation. Indeed, we found that c-Abl is required for proper activation of both Atm and Atr. c-Abl is bound to the chromatin and shows enhanced interaction with Atm and Atr in response to DNA damage. c-Abl can phosphorylate Atr on Y291 and Y310 and this phosphorylation appears to have a positive role in Atr activation under genotoxic stress. These findings suggest that Atm-mediated c-Abl activation in cell response to double-stranded DNA breaks might facilitate the activation of both Atm and Atr to regulate their downstream cellular events.
- Published
- 2011
- Full Text
- View/download PDF
30. Oxidative stress activates the c-Abl/p73 proapoptotic pathway in Niemann-Pick type C neurons.
- Author
-
Klein A, Maldonado C, Vargas LM, Gonzalez M, Robledo F, Perez de Arce K, Muñoz FJ, Hetz C, Alvarez AR, and Zanlungo S
- Subjects
- Animals, DNA-Binding Proteins metabolism, Disease Models, Animal, Down-Regulation drug effects, Down-Regulation physiology, Mice, Mice, Inbred BALB C, Neurons pathology, Niemann-Pick Disease, Type C genetics, Nuclear Proteins metabolism, Proto-Oncogene Proteins c-abl metabolism, Rats, Rats, Sprague-Dawley, Tumor Protein p73, Tumor Suppressor Proteins metabolism, Up-Regulation drug effects, Apoptosis physiology, DNA-Binding Proteins physiology, Neurons metabolism, Niemann-Pick Disease, Type C metabolism, Niemann-Pick Disease, Type C pathology, Nuclear Proteins physiology, Oxidative Stress physiology, Proto-Oncogene Proteins c-abl physiology, Tumor Suppressor Proteins physiology, Up-Regulation physiology
- Abstract
Niemann-Pick type C (NPC) is a neurodegenerative disease characterized by the intralysosomal accumulation of cholesterol leading to neuronal apoptosis. We have previously reported the activation of the c-Abl/p73 proapoptotic pathway in the cerebellum of NPC mice; however, upstream signals underlying the engagement of this pathway remain unknown. Here, we investigate the possible role of oxidative stress in the activation of c-Abl/p73 using different in vitro and in vivo NPC models. Our results indicate a close temporal correlation between the appearance of nitrotyrosine (N-Tyr; a post-translational tyrosine modification caused by oxidative stress) and the activation of c-Abl/p73 in NPC models. To test the functional role of oxidative stress in NPC, we have treated NPC neurons with the antioxidant NAC and observed a dramatic decrease of c-Abl/p73 activation and a reduction in the levels of apoptosis in NPC models. In conclusion, our data suggest that oxidative stress is the main upstream stimulus activating the c-Abl/p73 pathway and neuronal apoptosis in NPC neurons., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
31. Proteins that bind the Src homology 3 domain of CrkI have distinct roles in Crk transformation.
- Author
-
Zheng J, Machida K, Antoku S, Ng KY, Claffey KP, and Mayer BJ
- Subjects
- Animals, Apoptosis, Guanine Nucleotide-Releasing Factor 2 physiology, HEK293 Cells, Humans, Male, Mice, Mice, Nude, NIH 3T3 Cells, Proto-Oncogene Proteins c-abl antagonists & inhibitors, Proto-Oncogene Proteins c-abl physiology, Proto-Oncogene Proteins c-crk chemistry, SOS1 Protein physiology, Signal Transduction, Cell Transformation, Neoplastic, Proto-Oncogene Proteins c-crk physiology, src Homology Domains
- Abstract
The v-Crk oncogene product consists of two protein interaction modules, a Src homology 2 (SH2) domain and a Src homology 3 (SH3) domain. Overexpression of CrkI, the cellular homolog of v-Crk, transforms mouse fibroblasts, and elevated CrkI expression is observed in several human cancers. The SH2 and SH3 domains of Crk are required for transformation, but the identity of the critical cellular binding partners is not known. A number of candidate Crk SH3-binding proteins have been identified, including the nonreceptor tyrosine kinases c-Abl and Arg, and the guanine nucleotide exchange proteins C3G, SOS1 and DOCK180. The aim of this study is to determine which of these are required for transformation by CrkI. We found that short hairpin RNA-mediated knockdown of C3G or SOS1 suppressed anchorage-independent growth of NIH-3T3 cells overexpressing CrkI, whereas knockdown of SOS1 alone was sufficient to suppress tumor formation by these cells in nude mice. Knockdown of C3G was sufficient to revert morphological changes induced by CrkI expression. By contrast, knockdown of Abl family kinases or their inhibition with imatinib enhanced anchorage-independent growth and tumorigenesis induced by Crk. These results show that SOS1 is essential for CrkI-induced fibroblast transformation, and also reveal a surprising negative role for Abl kinases in Crk transformation.
- Published
- 2010
- Full Text
- View/download PDF
32. The Abl and Arg non-receptor tyrosine kinases regulate different zones of stress fiber, focal adhesion, and contractile network localization in spreading fibroblasts.
- Author
-
Peacock JG, Couch BA, and Koleske AJ
- Subjects
- 3T3 Cells, Actin Cytoskeleton metabolism, Actomyosin metabolism, Animals, Cell Adhesion physiology, Cytoskeleton metabolism, Fibroblasts cytology, Fibroblasts metabolism, Focal Adhesions enzymology, Mice, Myosin Light Chains metabolism, Stress Fibers enzymology, Fibroblasts physiology, Focal Adhesions physiology, Protein-Tyrosine Kinases physiology, Proto-Oncogene Proteins c-abl physiology, Stress Fibers physiology
- Abstract
Directed cell migration requires precise spatial control of F-actin-based leading edge protrusion, focal adhesion (FA) dynamics, and actomyosin contractility. In spreading fibroblasts, the Abl family kinases, Abl and Arg, primarily localize to the nucleus and cell periphery, respectively. Here we provide evidence that Abl and Arg exert different spatial regulation on cellular contractile and adhesive structures. Loss of Abl function reduces FA, F-actin, and phosphorylated myosin light chain (pMLC) staining at the cell periphery, shifting the distribution of these elements more to the center of the cell than in wild-type (WT) and arg(-/-) cells. Conversely, loss of Arg function shifts the distribution of these contractile and adhesion elements more to the cell periphery relative to WT and abl(-/-) cells. Abl/Arg-dependent phosphorylation of p190RhoGAP (p190) promotes its binding to p120RasGAP (p120) to form a functional RhoA GTPase inhibitory complex, which attenuates RhoA activity and downstream pMLC and FA formation. p120 and p190 colocalize both in the central region and at the cell periphery in WT cells. This p120:p190 colocalization redistributes to a more peripheral distribution in abl(-/-) cells and to a more centralized distribution in arg(-/-) cells, and these altered distributions can be restored to WT patterns via re-expression of Abl or Arg, respectively. Thus, the altered p120:p190 distribution in the mutant cells correlates inversely with the redistribution in adhesions, actin, and pMLC staining in these cells. Our studies suggest that Abl and Arg exert different spatial regulation on actomyosin contractility and focal adhesions within cells., (© 2010 Wiley-Liss, Inc.)
- Published
- 2010
- Full Text
- View/download PDF
33. Focus issue: Evolution III--domains for change.
- Author
-
Gough NR
- Subjects
- Animals, Humans, Protein Isoforms physiology, Protein Structure, Tertiary, Apoptosis physiology, Evolution, Molecular, Proto-Oncogene Proteins c-abl physiology, Signal Transduction physiology
- Abstract
This third installment in the series on the evolution of signaling molecules and networks highlights domains as a medium for evolution. Two Perspectives highlight how changes in domains can lead to new protein interactions or the evolution of new signaling events. Research in this issue highlights the evolution of proteins with domains involved in extrinsic apoptosis, a particular form of cell death caused by extracellular ligands that bind to "death receptors," and shows that this cell death pathway occurred earlier in evolution than was previously thought. A Review describes the evolution of the ABL family of tyrosine kinases, which have acquired isoform-specific functions through the unique combinations of domains and which are of particular importance in cancer.
- Published
- 2010
- Full Text
- View/download PDF
34. c-Abl, Lamellipodin, and Ena/VASP proteins cooperate in dorsal ruffling of fibroblasts and axonal morphogenesis.
- Author
-
Michael M, Vehlow A, Navarro C, and Krause M
- Subjects
- Animals, Blotting, Western, Cell Line, Drosophila melanogaster metabolism, Drosophila melanogaster physiology, Immunoprecipitation, Nerve Growth Factors physiology, Netrin-1, Phosphorylation, Platelet-Derived Growth Factor physiology, Tumor Suppressor Proteins physiology, Axons physiology, DNA-Binding Proteins physiology, Fibroblasts physiology, Morphogenesis physiology, Proto-Oncogene Proteins c-abl physiology
- Abstract
Background: Tight regulation of cell motility is essential for many physiological processes, such as formation of a functional nervous system and wound healing. Drosophila Abl negatively regulates the actin cytoskeleton effector protein Ena during neuronal development in flies, and it has been postulated that this may occur through an unknown intermediary. Lamellipodin (Lpd) regulates cell motility and recruits Ena/VASP proteins (Ena, Mena, VASP, EVL) to the leading edge of cells. However, the regulation of this recruitment has remained unsolved., Results: Here we show that Lpd is a substrate of Abl kinases and binds to the Abl SH2 domain. Phosphorylation of Lpd positively regulates the interaction between Lpd and Ena/VASP proteins. Consistently, efficient recruitment of Mena and EVL to Lpd at the leading edge requires Abl kinases. Furthermore, transient Lpd phosphorylation by Abl kinases upon netrin-1 stimulation of primary cortical neurons positively correlates with an increase in Lpd-Mena coprecipitation. Lpd is also transiently phosphorylated by Abl kinases upon platelet-derived growth factor (PDGF) stimulation, regulates PDGF-induced dorsal ruffling of fibroblasts and axonal morphogenesis, and cooperates with c-Abl in an Ena/VASP-dependent manner., Conclusions: Our findings suggest that Abl kinases positively regulate Lpd-Ena/VASP interaction, Ena/VASP recruitment to Lpd at the leading edge, and Lpd-Ena/VASP function in axonal morphogenesis and in PDGF-induced dorsal ruffling. Our data do not support the suggested negative regulatory role of Abl for Ena. Instead, we propose that Lpd is the hitherto unknown intermediary between Abl and Ena/VASP proteins., ((c) 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
35. c-Abl mediates endothelial apoptosis induced by inhibition of integrins alphavbeta3 and alphavbeta5 and by disruption of actin.
- Author
-
Xu J, Millard M, Ren X, Cox OT, and Erdreich-Epstein A
- Subjects
- Actin Cytoskeleton drug effects, Actin Cytoskeleton ultrastructure, Animals, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Cattle, Cell Adhesion, Cell Line, Tumor, Cell Shape drug effects, Culture Media pharmacology, Culture Media, Serum-Free, Endothelial Cells drug effects, Fetal Blood, Genes, abl, Glioblastoma pathology, Humans, Medulloblastoma pathology, Membrane Potential, Mitochondrial, Neuroblastoma pathology, Proto-Oncogene Proteins c-abl antagonists & inhibitors, RNA Interference, RNA, Small Interfering genetics, Thiazolidines pharmacology, Actins antagonists & inhibitors, Apoptosis physiology, Endothelial Cells cytology, Integrin alphaVbeta3 antagonists & inhibitors, Proto-Oncogene Proteins c-abl physiology, Receptors, Vitronectin antagonists & inhibitors
- Abstract
Inhibition of integrins alphavbeta3 and alphavbeta5 in human brain microvascular endothelial cells (HBMECs) by the function-blocking peptide RGDfV induces loss of spreading on vitronectin, cell detachment, and apoptosis. We demonstrate that cell detachment is not required for apoptosis because plating on bovine serum albumin-blocked poly-L-lysine (allows attachment, but not integrin ligation and cell spreading) also induced apoptosis. Latrunculin B (LatB), which inhibits F-actin polymerization, induced transient loss of HBMEC spreading on vitronectin, but not their detachment, and induced apoptosis despite recovery of cell spreading. However, LatB did not cause apoptosis in 5 tumor cell lines. In HBMECs, both LatB and RGDfV induced transient Y412 and Y245 phosphorylation of endogenous c-Abl, a nonreceptor tyrosine kinase that reciprocally regulates F-actin. LatB also induced nuclear translocation of c-Abl in HBMECs. STI-571 (imatinib), a targeted therapy for BCR-ABL1(+) leukemias and inhibitor of c-Abl, platelet-derived growth factor receptor, and c-Kit, decreased endothelial apoptosis. LatB-induced HBMEC apoptosis, and its inhibition by STI-571 also occurred in a 3-dimensional collagen model, supporting physiologic relevance. Last, siRNA to c-Abl (but not nonspecific siRNA) also inhibited RGDfV- and LatB-induced apoptosis. Thus, endogenous c-Abl mediates endothelial apoptosis induced by inhibition of integrins alphavbeta3/alphavbeta5 or by LatB-induced disruption of F-actin.
- Published
- 2010
- Full Text
- View/download PDF
36. Synaptic clustering of PSD-95 is regulated by c-Abl through tyrosine phosphorylation.
- Author
-
Perez de Arce K, Varela-Nallar L, Farias O, Cifuentes A, Bull P, Couch BA, Koleske AJ, Inestrosa NC, and Alvarez AR
- Subjects
- Animals, Biomarkers metabolism, Cell Line, Cells, Cultured, Disks Large Homolog 4 Protein, Humans, Male, Membrane Proteins ultrastructure, Mice, Mice, Knockout, Phosphorylation physiology, Proto-Oncogene Proteins c-abl ultrastructure, Rats, Rats, Sprague-Dawley, Synapses ultrastructure, Intracellular Signaling Peptides and Proteins metabolism, Membrane Proteins metabolism, Proto-Oncogene Proteins c-abl physiology, Synapses metabolism, Tyrosine metabolism
- Abstract
The c-Abl tyrosine kinase is present in mouse brain synapses, but its precise synaptic function is unknown. We found that c-Abl levels in the rat hippocampus increase postnatally, with expression peaking at the first postnatal week. In 14 d in vitro hippocampal neuron cultures, c-Abl localizes primarily to the postsynaptic compartment, in which it colocalizes with the postsynaptic scaffold protein postsynaptic density protein-95 (PSD-95) in apposition to presynaptic markers. c-Abl associates with PSD-95, and chemical or genetic inhibition of c-Abl kinase activity reduces PSD-95 tyrosine phosphorylation, leading to reduced PSD-95 clustering and reduced synapses in treated neurons. c-Abl can phosphorylate PSD-95 on tyrosine 533, and mutation of this residue reduces the ability of PSD-95 to cluster at postsynaptic sites. Our results indicate that c-Abl regulates synapse formation by mediating tyrosine phosphorylation and clustering of PSD-95.
- Published
- 2010
- Full Text
- View/download PDF
37. N-myristoylated c-Abl tyrosine kinase localizes to the endoplasmic reticulum upon binding to an allosteric inhibitor.
- Author
-
Choi Y, Seeliger MA, Panjarian SB, Kim H, Deng X, Sim T, Couch B, Koleske AJ, Smithgall TE, and Gray NS
- Subjects
- 3T3 Cells, Adenosine Triphosphate chemistry, Allosteric Site, Animals, Antineoplastic Agents pharmacology, Binding Sites, Drug Resistance, Neoplasm, Humans, Lipids chemistry, Mice, Molecular Conformation, Protein Isoforms, Protein Structure, Tertiary, Proto-Oncogene Proteins c-abl metabolism, Endoplasmic Reticulum enzymology, Proto-Oncogene Proteins c-abl physiology
- Abstract
Allosteric kinase inhibitors hold promise for revealing unique features of kinases that may not be apparent using conventional ATP-competitive inhibitors. Here we explore the activity of a previously reported allosteric inhibitor of BCR-Abl kinase, GNF-2, against two cellular isoforms of Abl tyrosine kinase: one that carries a myristate in the N terminus and the other that is deficient in N-myristoylation. Our results show that GNF-2 inhibits the kinase activity of non-myristoylated c-Abl more potently than that of myristoylated c-Abl by binding to the myristate-binding pocket in the C-lobe of the kinase domain. Unexpectedly, indirect immunofluorescence reveals a translocation of myristoylated c-Abl to the endoplasmic reticulum in GNF-2-treated cells, whereas GNF-2 has no detectable effect on the localization of non-myristoylated c-Abl. These results indicate that GNF-2 competes with the NH(2)-terminal myristate for binding to the c-Abl kinase myristate-binding pocket and that the exposed myristoyl group accounts for the localization to the endoplasmic reticulum. We also demonstrate that GNF-2 can inhibit enzymatic and cellular kinase activity of Arg, a kinase highly homologous to c-Abl, which is also likely to be regulated through intramolecular binding of an NH(2)-terminal myristate lipid. These results suggest that non-ATP-competitive inhibitors, such as GNF-2, can serve as chemical tools that can discriminate between c-Abl isoform-specific behaviors.
- Published
- 2009
- Full Text
- View/download PDF
38. The effects of c-Abl mutation on developing B cell differentiation and survival.
- Author
-
Brightbill H and Schlissel MS
- Subjects
- Animals, Antigens, CD19 immunology, Antigens, CD19 metabolism, Apoptosis genetics, Calcium immunology, Calcium metabolism, Cell Proliferation drug effects, Cell Survival genetics, Interleukin-7 pharmacology, Mice, Mice, Knockout, Mutation, Precursor Cells, B-Lymphoid drug effects, Precursor Cells, B-Lymphoid enzymology, Proto-Oncogene Proteins c-abl genetics, Recombinant Proteins pharmacology, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 immunology, Tumor Suppressor Protein p53 metabolism, Cell Differentiation genetics, Precursor Cells, B-Lymphoid immunology, Proto-Oncogene Proteins c-abl physiology
- Abstract
c-Abl is a widely expressed Src family protein tyrosine kinase that is activated by chromosomal translocation in certain human leukemias. While shown in various experimental systems to regulate cell division and stress responses, its biological functions remain poorly understood. Although expressed at similar levels throughout B cell development, we found that the fraction of phosphorylated, active c-Abl peaks at the pro-B stage. We went on to perform a detailed analysis of B cell development in c-Abl-deficient mice. We confirmed a striking but variable decrease in pro- and pre-B cell numbers, a decrease in pre-B cell growth and an increase in pre-B cell apoptosis. This phenotype was not rescued by transgenic expression of a functional IgHC transgene and only partially rescued by the anti-apoptosis gene Bcl-x. Unlike their wild-type counterparts, c-Abl-deficient pre-B cells show a defect in Ca(2+) flux upon cross-linking of CD19, a co-receptor known to be involved in pre-B cell receptor signaling and failed to express CD25 on the cell surface. Despite these pre-B cell-signaling defects, selection for in-frame heavy-chain rearrangements was intact in the mutant mice. Remarkably, we were able to rescue the proliferative defect by culturing cells in vitro with large amounts of rIL-7. We conclude that c-Abl is required for normal B cell differentiation and survival.
- Published
- 2009
- Full Text
- View/download PDF
39. A non-Smad mechanism of fibroblast activation by transforming growth factor-beta via c-Abl and Egr-1: selective modulation by imatinib mesylate.
- Author
-
Bhattacharyya S, Ishida W, Wu M, Wilkes M, Mori Y, Hinchcliff M, Leof E, and Varga J
- Subjects
- Animals, Benzamides, Bleomycin toxicity, Cells, Cultured, Collagen genetics, Extracellular Signal-Regulated MAP Kinases physiology, Fibroblasts physiology, Fibrosis, Humans, Imatinib Mesylate, Mice, Mice, Inbred BALB C, NIH 3T3 Cells, Signal Transduction, Smad2 Protein physiology, Smad3 Protein physiology, Early Growth Response Protein 1 physiology, Fibroblasts drug effects, Piperazines pharmacology, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-abl physiology, Pyrimidines pharmacology, Transforming Growth Factor beta pharmacology
- Abstract
The nonreceptor protein tyrosine kinase c-Abl regulates cell proliferation and survival. Recent studies provide evidence that implicate c-Abl as a mediator for fibrotic responses induced by transforming growth factor-beta (TGF-beta), but the precise mechanisms underlying this novel oncogene function are unknown. Here, we report that when expressed in normal fibroblasts, a constitutively active mutant of Abl that causes chronic myelogenous leukemia (CML) stimulated the expression and transcriptional activity of the early growth response factor 1 (Egr-1). Mouse embryonic fibroblasts (MEFs), lacking c-Abl, were resistant to TGF-beta stimulation. Responsiveness of these MEFs to TGF-beta could be rescued by wild-type c-Abl, but not by a kinase-deficient mutant form of c-Abl. Furthermore, Abl kinase activity was necessary for the induction of Egr-1 by TGF-beta in normal fibroblasts, and Egr-1 was required for stimulation of collagen by Bcr-Abl. Lesional skin fibroblasts in mice with bleomycin-induced fibrosis of skin displayed evidence of c-Abl activation in situ, and elevated phospho-c-Abl correlated with increased local expression of Egr-1. Collectively, these results position Egr-1 downstream of c-Abl in the fibrotic response, delineate a novel Egr-1-dependent intracellular signaling mechanism that underlies the involvement of c-Abl in certain TGF-beta responses, and identify Egr-1 as a target of inhibition by imatinib. Furthermore, the findings show in situ activation of c-Abl paralleling the upregulated tissue expression of Egr-1 that accompanies fibrosis. Pharmacological targeting of c-Abl and its downstream effector pathways may, therefore, represent a novel therapeutic approach to blocking TGF-beta-dependent fibrotic processes.
- Published
- 2009
- Full Text
- View/download PDF
40. c-Abl kinase is required for beta 2 integrin-mediated neutrophil adhesion.
- Author
-
Cui L, Chen C, Xu T, Zhang J, Shang X, Luo J, Chen L, Ba X, and Zeng X
- Subjects
- CD18 Antigens metabolism, Cell Adhesion immunology, Cytoplasm enzymology, Cytoplasm immunology, Cytoplasm metabolism, HL-60 Cells, Humans, Neutrophil Activation immunology, Neutrophils enzymology, Protein Structure, Tertiary, Proto-Oncogene Proteins c-abl metabolism, Signal Transduction immunology, Talin physiology, Tumor Necrosis Factor-alpha physiology, CD18 Antigens physiology, Neutrophils cytology, Neutrophils immunology, Proto-Oncogene Proteins c-abl physiology
- Abstract
Integrin regulation in neutrophil adhesion is essential for innate immune response. c-Abl kinase is a nonreceptor tyrosine kinase and is critical for signaling transduction from various receptors in leukocytes. Using neutrophils and dHL-60 (neutrophil-like differentiation of HL-60) cells, we show that c-Abl kinase is activated by beta(2) integrin engagement and is required for beta(2) integrin-dependent neutrophil sustained adhesion and spreading. The expression of beta(2) integrin on neutrophils induced by TNF-alpha is not affected by c-Abl kinase inhibitor STI571, suggesting that c-Abl kinase is not involved in TNF-alpha-induced integrin activation. The recruitment of c-Abl kinase to beta(2) integrin is dependent on talin head domain, which constitutively interacts with beta(2) integrin cytoplasmic domain. After activated, c-Abl kinase increases the tyrosine phosphorylation of Vav. The SH3 domain of c-Abl kinase is involved in its interaction with talin and Vav. Thus, c-Abl kinase plays an essential role in the activation of Vav induced by beta(2) integrin ligation and in regulating neutrophil-sustained adhesion and spreading.
- Published
- 2009
- Full Text
- View/download PDF
41. Abelson tyrosine kinase facilitates Salmonella enterica serovar Typhimurium entry into epithelial cells.
- Author
-
Ly KT and Casanova JE
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Benzamides, Cytoskeletal Proteins metabolism, HeLa Cells, Humans, Imatinib Mesylate, Phosphorylation, Piperazines pharmacology, Protein Kinase Inhibitors pharmacology, Protein-Tyrosine Kinases antagonists & inhibitors, Protein-Tyrosine Kinases physiology, Proto-Oncogene Proteins c-abl antagonists & inhibitors, Proto-Oncogene Proteins c-crk metabolism, Pyrimidines pharmacology, Epithelial Cells microbiology, Proto-Oncogene Proteins c-abl physiology, Salmonella typhi pathogenicity
- Abstract
The intracellular gram-negative bacterial pathogen Salmonella enterica serovar Typhimurium gains entry into nonphagocytic cells by manipulating the assembly of the host actin cytoskeleton. S. enterica serovar Typhimurium entry requires a functional type III secretion system, a conduit through which bacterial effector proteins are directly translocated into the host cytosol. We and others have previously reported the enhancement of tyrosine kinase activities during Salmonella serovar Typhimurium infection; however, neither specific kinases nor their targets have been well characterized. In this study, we investigated the roles of the cellular Abelson tyrosine kinase (c-Abl) and the related protein Arg in the context of serovar Typhimurium infection. We found that bacterial internalization was inhibited by more than 70% in cells lacking both c-Abl and Arg and that treatment of wild-type cells with a pharmaceutical inhibitor of the c-Abl kinase, STI571 (imatinib), reduced serovar Typhimurium invasion efficiency to a similar extent. Bacterial infection led to enhanced phosphorylation of two previously identified c-Abl substrates, the adaptor protein CT10 regulator of kinase (CrkII) and the Abelson-interacting protein Abi1, a component of the WAVE2 complex. Furthermore, overexpression of the nonphosphorylatable form of CrkII resulted in decreased invasion. Taken together, these findings indicate that c-Abl is activated during S. enterica serovar Typhimurium infection and that its phosphorylation of multiple downstream targets is functionally important in bacterial internalization.
- Published
- 2009
- Full Text
- View/download PDF
42. Novel N9-arenethenyl purines as potent dual Src/Abl tyrosine kinase inhibitors.
- Author
-
Wang Y, Shakespeare WC, Huang WS, Sundaramoorthi R, Lentini S, Das S, Liu S, Banda G, Wen D, Zhu X, Xu Q, Keats J, Wang F, Wardwell S, Ning Y, Snodgrass JT, Broudy MI, Russian K, Dalgarno D, Clackson T, and Sawyer TK
- Subjects
- Animals, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Cell Proliferation drug effects, Growth Inhibitors chemistry, Growth Inhibitors pharmacology, Humans, K562 Cells, Protein Kinase Inhibitors chemistry, Proto-Oncogene Proteins c-abl physiology, Rats, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-abl antagonists & inhibitors, Purines chemistry, Purines pharmacology, src-Family Kinases antagonists & inhibitors
- Abstract
Novel N(9)-arenethenyl purines, optimized potent dual Src/Abl tyrosine kinase inhibitors, are described. The key structural feature is a trans vinyl linkage at N(9) on the purine core which projects hydrophobic substituents into the selectivity pocket at the rear of the ATP site. Their synthesis was achieved through a Horner-Wadsworth-Emmons reaction of N(9)-phosphorylmethylpurines and substituted benzaldehydes or Heck reactions between 9-vinyl purines and aryl halides. Most compounds are potent inhibitors of both Src and Abl kinase, and several possess good oral bioavailability.
- Published
- 2008
- Full Text
- View/download PDF
43. Inhibition of Abl tyrosine kinase enhances nerve growth factor-mediated signaling in Bcr-Abl transformed cells via the alteration of signaling complex and the receptor turnover.
- Author
-
Koch A, Scherr M, Breyer B, Mancini A, Kardinal C, Battmer K, Eder M, and Tamura T
- Subjects
- Animals, Benzamides, Cell Line, Tumor, Cell Transformation, Neoplastic metabolism, Extracellular Signal-Regulated MAP Kinases metabolism, Fusion Proteins, bcr-abl, Gene Silencing physiology, Humans, Imatinib Mesylate, K562 Cells, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Leukemia, Myelogenous, Chronic, BCR-ABL Positive metabolism, Multiprotein Complexes metabolism, PC12 Cells, Phosphorylation drug effects, Protein Kinase Inhibitors pharmacology, Protein-Tyrosine Kinases antagonists & inhibitors, Proto-Oncogene Proteins c-abl metabolism, Proto-Oncogene Proteins c-abl physiology, Proto-Oncogene Proteins c-akt metabolism, RNA, Small Interfering pharmacology, Rats, Signal Transduction drug effects, Cell Transformation, Neoplastic genetics, Nerve Growth Factor pharmacology, Piperazines pharmacology, Protein-Tyrosine Kinases genetics, Proto-Oncogene Proteins c-abl antagonists & inhibitors, Pyrimidines pharmacology, Receptor, trkA metabolism
- Abstract
Receptor tyrosine kinase-mediated signaling is tightly regulated by a number of cytoplasmic signaling molecules. In this report, we show that Bcr-Abl transformed chronic myelogenous leukemia (CML) cell lines, K562 and Meg-01, express the receptor for nerve growth factor (NGF), TrkA, on the cell surface; however, the NGF-mediated signal is not particularly strong. Treatment with imatinib, a potent inhibitor of Bcr-Abl tyrosine kinase, downmodulates phosphorylation of downstream molecules. Upon stimulation with NGF, Erk and Akt are phosphorylated to a much greater degree in imatinib-treated cells than in untreated cells. Knockdown of expression of Bcr-Abl using small interfering RNA technique also enhanced NGF-mediated Akt phosphorylation, indicating that Bcr-Abl kinase modifies NGF signaling directly. Imatinib treatment also enhanced NGF signaling in rat adrenal pheochromocytoma cell line PC12 that expresses TrkA and c-Abl, suggesting that it is not only restoration of responsiveness to NGF after blocking oncoprotein activity, but also c-Abl tyrosine kinase per se may be a negative regulator of growth factor signaling. Furthermore, inhibition of Abl tyrosine kinase enhanced clearance of surface TrkA after NGF treatment and simultaneously enhanced NGF-mediated signaling, suggesting that as in neuronal cells 'signaling endosomes' are formed in hematopoietic cells. To examine the role of TrkA in CML cells, we studied cell growth or colony formation in the presence or absence of imatinib with or without NGF. We found that NGF treatment induces cell survival in imatinib-treated CML cell lines, as well as colony formation of primary CD34+ CML cells, strongly suggesting that NGF/TrkA signaling contributes to aberrant signaling in CML.
- Published
- 2008
- Full Text
- View/download PDF
44. Role of c-Abl kinase in DNA mismatch repair-dependent G2 cell cycle checkpoint arrest responses.
- Author
-
Wagner MW, Li LS, Morales JC, Galindo CL, Garner HR, Bornmann WG, and Boothman DA
- Subjects
- Antineoplastic Agents pharmacology, Base Pair Mismatch, Benzamides, Cell Cycle, Cell Cycle Proteins metabolism, Cell Line, Tumor, Colonic Neoplasms drug therapy, Colonic Neoplasms metabolism, Dose-Response Relationship, Drug, G2 Phase, Humans, Imatinib Mesylate, Methylnitronitrosoguanidine pharmacology, Models, Biological, Nuclear Proteins metabolism, Piperazines, Proto-Oncogene Proteins c-abl metabolism, Pyrimidines pharmacology, Signal Transduction, DNA Repair, Proto-Oncogene Proteins c-abl physiology
- Abstract
Current published data suggest that DNA mismatch repair (MMR) triggers prolonged G(2) cell cycle checkpoint arrest after alkylation damage from N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) by activating ATR (ataxia telangiectasia-Rad3-related kinase). However, analyses of isogenic MMR-proficient and MMR-deficient human RKO colon cancer cells revealed that although ATR/Chk1 signaling controlled G(2) arrest in MMR-deficient cells, ATR/Chk1 activation was not involved in MMR-dependent G(2) arrest. Instead, we discovered that disrupting c-Abl activity using STI571 (Gleevec, a c-Abl inhibitor) or stable c-Abl knockdown abolished MMR-dependent p73alpha stabilization, induction of GADD45alpha protein expression, and G(2) arrest. In addition, inhibition of c-Abl also increased the survival of MNNG-exposed MMR-proficient cells to a level comparable with MMR-deficient cells. Furthermore, knocking down GADD45alpha (but not p73alpha) protein levels affected MMR-dependent G(2) arrest responses. Thus, MMR-dependent G(2) arrest responses triggered by MNNG are dependent on a human MLH1/c-Abl/GADD45alpha signaling pathway and activity. Furthermore, our data suggest that caution should be taken with therapies targeting c-Abl kinase because increased survival of mutator phenotypes may be an unwanted consequence.
- Published
- 2008
- Full Text
- View/download PDF
45. c-Abl is involved in high glucose-induced apoptosis in embryonic E12.5 cortical neural progenitor cells from the mouse brain.
- Author
-
Jia DY, Du ZH, Liu SM, Liu HJ, Wang FW, Ling EA, Liu K, and Hao AJ
- Subjects
- Animals, Apoptosis drug effects, Cell Differentiation drug effects, Cell Differentiation physiology, Cells, Cultured, Cerebral Cortex embryology, Glucose administration & dosage, Mice, Mice, Mutant Strains, Neurons cytology, Neurons metabolism, Proto-Oncogene Proteins c-abl biosynthesis, Proto-Oncogene Proteins c-abl genetics, Reactive Oxygen Species metabolism, Signal Transduction drug effects, Signal Transduction physiology, Stem Cells cytology, Stem Cells metabolism, Apoptosis physiology, Cerebral Cortex cytology, Cerebral Cortex metabolism, Glucose toxicity, Neurons physiology, Proto-Oncogene Proteins c-abl physiology, Stem Cells physiology
- Abstract
Hyperglycemia causes direct apoptosis of neural progenitor cells (NPCs) in diabetic-induced neural tube defects in embryos. However, the underlying mechanisms are poorly understood. The present study is aimed to investigate the specific cellular proteins that may be involved in NPCs apoptosis as well as mechanisms by which the proteins regulate the oxidative stress-induced NPCs apoptosis. Our present results have shown that the expression of c-Abl was up-regulated in NPCs exposed to high glucose in vitro. The increased c-Abl was localized mainly in the nucleus. High glucose also induced an increase in nuclear p53 protein levels and the p53-c-Abl complex in NPCs. Administration of reactive oxygen species scavengers decreased the protein level of c-Abl, p53 and NPCs apoptosis. Inhibition of c-Abl reduced NPCs apoptosis and the nuclear protein level of p53 in response to high glucose. These results demonstrate that c-Abl is involved in the reactive oxygen species-activated apoptotic pathways in NPCs apoptosis. Inhibition of c-Abl may protect NPCs against insults induced by high glucose via the modulation of NPCs apoptotic machinery.
- Published
- 2008
- Full Text
- View/download PDF
46. Activated c-Abl tyrosine kinase in malignant solid tumors.
- Author
-
Lin J and Arlinghaus R
- Subjects
- Breast Neoplasms etiology, Breast Neoplasms pathology, Carcinoma, Non-Small-Cell Lung etiology, Carcinoma, Non-Small-Cell Lung pathology, Cell Line, Tumor, Cell Proliferation, Enzyme Activation, ErbB Receptors physiology, Humans, Lung Neoplasms etiology, Lung Neoplasms pathology, Proto-Oncogene Proteins c-abl genetics, Proto-Oncogenes, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins physiology, Breast Neoplasms enzymology, Carcinoma, Non-Small-Cell Lung enzymology, Lung Neoplasms enzymology, Proto-Oncogene Proteins c-abl physiology
- Abstract
Mutant forms of the c-ABL gene are well known to be involved in hematopoietic malignancies such as chronic myeloid leukemia (CML). CML patients possess a fused BCR-ABL gene that activates the Abl tyrosine kinase domain within Bcr-Abl. In general fusion proteins that cause oligomerization of Abl lead to activation of its tyrosine kinase activity. In this review, we highlight recent discoveries indicating that the activated c-Abl tyrosine kinase, not as a fusion protein, plays an important role in malignant solid tumors of lung and breast.
- Published
- 2008
- Full Text
- View/download PDF
47. Pharmacological inhibition of c-Abl compromises genetic stability and DNA repair in Bcr-Abl-negative cells.
- Author
-
Fanta S, Sonnenberg M, Skorta I, Duyster J, Miething C, Aulitzky WE, and van der Kuip H
- Subjects
- Animals, Antineoplastic Agents pharmacology, Benzamides, Chromosome Aberrations, DNA Damage, Dasatinib, Fibroblasts metabolism, Humans, Imatinib Mesylate, Kinetics, Leukocytes, Mononuclear drug effects, Leukocytes, Mononuclear radiation effects, Mice, Piperazines pharmacology, Proto-Oncogene Proteins c-abl antagonists & inhibitors, Pyrimidines pharmacology, Thiazoles pharmacology, DNA Repair, Fusion Proteins, bcr-abl biosynthesis, Gene Expression Regulation, Neoplastic, Proto-Oncogene Proteins c-abl physiology
- Abstract
Imatinib inhibits the kinase activity of Bcr-Abl and is currently the most effective drug for treatment of chronic myeloid leukemia (CML). Imatinib also blocks c-Abl, a physiological tyrosine kinase activated by a variety of stress signals including damaged DNA. We investigated the effect of pharmacological inhibition of c-Abl on the processing of irradiation-induced DNA damage in Bcr-Abl-negative cells. Cell lines and peripheral blood mononuclear cells (PBMCs) from healthy volunteers were treated with imatinib or dasatinib before gamma-irradiation. Inhibition of c-Abl caused an enhanced irradiation-induced mutation frequency and slowdown of DNA repair, whereas imatinib was ineffective in cells expressing a T315I variant of c-Abl. Mutation frequency and repair kinetics were also studied in c-Abl-/- murine embryonic fibroblasts (MEFs) retransfected with wild-type c-Abl (wt-Abl) or a kinase-defect variant of Abl (KD-Abl). Enhanced mutation frequency as well as delayed DNA repair was observed in cells expressing KD-Abl. These data indicate that pharmacological inhibition of c-Abl compromises DNA-damage response.
- Published
- 2008
- Full Text
- View/download PDF
48. [Mechanisms for induction of apoptosis in response to DNA damage].
- Author
-
Yoshida K
- Subjects
- 14-3-3 Proteins physiology, Active Transport, Cell Nucleus, Animals, Ataxia Telangiectasia Mutated Proteins, Cell Cycle genetics, Cell Cycle physiology, Cell Cycle Proteins genetics, Cell Cycle Proteins physiology, DNA-Binding Proteins genetics, DNA-Binding Proteins physiology, Genes, cdc physiology, Humans, JNK Mitogen-Activated Protein Kinases physiology, Protein Kinase C-delta physiology, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases physiology, Protein-Tyrosine Kinases physiology, Proto-Oncogene Proteins c-abl physiology, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins physiology, Dyrk Kinases, Apoptosis genetics, Apoptosis physiology, DNA Damage genetics, DNA Damage physiology, Signal Transduction genetics, Signal Transduction physiology
- Published
- 2008
49. Systems analysis of quantitative shRNA-library screens identifies regulators of cell adhesion.
- Author
-
Huang X, Wang JY, and Lu X
- Subjects
- Cell Line, Cell Separation, Cytokine Receptor gp130 physiology, Databases, Genetic, Gene Expression Profiling methods, Humans, Proto-Oncogene Proteins c-abl physiology, Reproducibility of Results, Semantics, Signal Transduction, Systems Analysis, Systems Biology methods, Transcription, Genetic physiology, Cell Adhesion genetics, Gene Library, RNA Interference physiology, RNA, Double-Stranded analysis
- Abstract
Background: High throughput screens with RNA interference technology enable loss-of-function analyses of gene activities in mammalian cells. While the construction of genome-scale shRNA libraries has been successful, results of large-scale screening of those libraries can be difficult to analyze because of the relatively high noise levels and the fact that not all shRNAs in a library are equally effective in silencing gene expression., Results: We have screened a library consisting of 43,828 shRNAs directed against 8,500 human genes for functions that are necessary in cell detachment induced by a constitutively activated c-Abl tyrosine kinase. To deal with the issues of noise and uncertainty of knockdown efficiencies, we employed an analytical strategy that combines quantitative data analysis with biological knowledge, i.e. Gene Ontology and pathway information, to increase the power of the RNAi screening technique. Using this strategy we found 16 candidate genes to be involved in Abl-induced disruption of cell adhesion, and verified that the knockdown of IL6ST is associated with enhanced cell attachment., Conclusion: Our results suggest that the power of genome-wide quantitative shRNA screens can be significantly increased when analyzed using a systems biology-based approach to identify functional gene networks.
- Published
- 2008
- Full Text
- View/download PDF
50. In vitro biotransformation of imatinib by the tumor expressed CYP1A1 and CYP1B1.
- Author
-
Rochat B, Zoete V, Grosdidier A, von Grünigen S, Marull M, and Michielin O
- Subjects
- Benzamides, Biotransformation, Cytochrome P-450 CYP1B1, Humans, Imatinib Mesylate, Kinetics, Mass Spectrometry, Proto-Oncogene Proteins c-abl physiology, Antineoplastic Agents pharmacokinetics, Aryl Hydrocarbon Hydroxylases physiology, Cytochrome P-450 CYP1A1 physiology, Neoplasms metabolism, Piperazines pharmacokinetics, Pyrimidines pharmacokinetics
- Abstract
The main objective of the study was to examine the biotransformation of the anticancer drug imatinib in target cells by incubating it with oxidoreductases expressed in tumor cells. The second objective was to obtain an in silico prediction of the potential activity of imatinib metabolites. An in vitro enzyme kinetic study was performed with cDNA expressed human oxidoreductases and LC-MS/MS analysis. The kinetic parameters (Km and Vmax) were determined for six metabolites. A molecular modeling approach was used to dock these metabolites to the target Abl or Bcr-Abl kinases. CYP3A4 isozyme showed the broadest metabolic capacity, whereas CYP1A1, CYP1B1 and FMO3 isozymes biotransformed imatinib with a high intrinsic clearance. The predicted binding modes for the metabolites to Abl were comparable to that of the parent drug, suggesting potential activity. These findings indicate that CYP1A1 and CYP1B1, which are known to be overexpressed in a wide range of tumors, are involved in the biotransformation of imatinib. They could play a role in imatinib disposition in the targeted stem, progenitor and differentiated cancer cells, with a possible contribution of the metabolites toward the activity of the drug., (Copyright (c) 2008 John Wiley & Sons, Ltd.)
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.