Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 158
Filter
2.
Biochim Biophys Acta Mol Basis Dis ; 1867(5): 166089, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33549745

ABSTRACT

Gaucher disease (GD) is caused by homozygous mutations in the GBA1 gene, which encodes the lysosomal ß-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.


Subject(s)
Apoptosis , Gaucher Disease/pathology , Glucosylceramidase/metabolism , Neurons/pathology , Proto-Oncogene Proteins c-abl/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , 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
3.
J Acquir Immune Defic Syndr ; 82(4): 407-415, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31658184

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.


Subject(s)
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 , 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
4.
CNS Neurol Disord Drug Targets ; 17(1): 14-21, 2018 04 26.
Article in English | MEDLINE | ID: mdl-28571531

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.


Subject(s)
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 , Animals , Humans , Neuroprotection/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
5.
J Exp Med ; 214(4): 1049-1064, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28289053

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.


Subject(s)
Capillary Permeability , Neuropilin-1/physiology , Proto-Oncogene Proteins c-abl/physiology , Vascular Endothelial Growth Factor A/physiology , src-Family Kinases/metabolism , Adaptor Proteins, Signal Transducing/physiology , Animals , Enzyme Activation , Mice , Mice, Inbred C57BL , Semaphorin-3A/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology
6.
Blood ; 127(17): 2131-43, 2016 04 28.
Article in English | MEDLINE | ID: mdl-26864341

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.


Subject(s)
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 , 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
7.
Neuromolecular Med ; 17(3): 297-304, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25989848

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.


Subject(s)
Autism Spectrum Disorder/etiology , Neurites/pathology , Neurogenesis , 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
8.
Oncotarget ; 6(11): 8851-74, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25860930

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.


Subject(s)
Cell Cycle Proteins/physiology , Intracellular Signaling Peptides and Proteins/physiology , Neoplasm Proteins/physiology , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Signal Transduction/physiology , 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
9.
Clin Sci (Lond) ; 128(1): 17-28, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24865476

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 ß-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 ß-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-ßH1 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 ß-cell function and that LRP1 is adversely affected by challenges to ß-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 ß-cells.


Subject(s)
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 , 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
10.
Proc Natl Acad Sci U S A ; 111(46): 16365-70, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25368164

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.


Subject(s)
Adipocytes/metabolism , Adipogenesis/physiology , PPAR gamma/physiology , Proto-Oncogene Proteins c-abl/physiology , 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
11.
Mol Cell ; 56(1): 163-73, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25219501

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 AßO-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.


Subject(s)
Alzheimer Disease/genetics , Histone Deacetylase 2/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-abl/physiology , Epigenesis, Genetic , Gene Expression Regulation , HeLa Cells , Humans , Phosphorylation , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Tyrosine/metabolism
12.
J Biol Chem ; 289(31): 21463-72, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24923444

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 ßc 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.


Subject(s)
Bone Marrow Cells/enzymology , Janus Kinase 2/metabolism , Proto-Oncogene Proteins c-abl/physiology , 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
13.
PLoS One ; 9(5): e97694, 2014.
Article in English | MEDLINE | ID: mdl-24835010

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 ß cells to promote insulin production. In this study, we showed that Imatinib significantly induced insulin expression in both glucose-stimulated and resting ß cells. In line with this finding, c-Abl knockdown by siRNA and overexpression of c-Abl markedly enhanced and inhibited insulin expression in ß 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 ß 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 ß cells. Our study demonstrates a previously unrecognized mechanism that controls insulin expression through c-Abl-regulated NKx2.2 and GLUT2. Therapeutic targeting ß cell c-Abl could be employed in the treatment of diabetes or ß cell tumor, insulinoma.


Subject(s)
Benzamides/pharmacology , Glucose Transporter Type 2/metabolism , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/drug effects , Insulin/biosynthesis , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Pyrimidines/pharmacology , Transcription Factors/metabolism , 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
14.
Neuromolecular Med ; 16(2): 217-30, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24522549

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.


Subject(s)
Oxidative Stress , Parkinson Disease/etiology , 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
15.
Mol Cell Biol ; 34(7): 1188-97, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24421390

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.


Subject(s)
Genes, abl , Oncogene Proteins v-abl/physiology , Proto-Oncogene Proteins c-abl/physiology , 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
16.
Respir Res ; 14: 105, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-24112389

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.


Subject(s)
Airway Remodeling/physiology , Asthma/physiopathology , Bronchial Hyperreactivity/physiopathology , Myocytes, Smooth Muscle/physiology , Proto-Oncogene Proteins c-abl/physiology , 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
17.
Biochem Biophys Res Commun ; 440(2): 197-203, 2013 Oct 18.
Article in English | MEDLINE | ID: mdl-23985350

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.


Subject(s)
Intracellular Signaling Peptides and Proteins/pharmacology , Myoblasts, Cardiac/drug effects , Myocardial Reperfusion Injury/prevention & control , Oxidative Stress/drug effects , Peptides/pharmacology , 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
18.
Nat Rev Cancer ; 13(8): 559-71, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23842646

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.


Subject(s)
Carcinoma/etiology , Carcinoma/therapy , Leukemia/etiology , Leukemia/therapy , Proto-Oncogene Proteins c-abl/physiology , Carcinoma/enzymology , Humans , Leukemia/enzymology , Protein Kinase Inhibitors/therapeutic use , Signal Transduction
19.
Cell Death Differ ; 20(7): 953-62, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23660976

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.


Subject(s)
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 , 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
20.
J Hepatol ; 57(6): 1292-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22889954

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.


Subject(s)
Hepatocytes/physiology , Liver/embryology , Proto-Oncogene Proteins c-abl/physiology , Proto-Oncogene Proteins c-met/physiology , Transcription, Genetic , Tumor Suppressor Protein p53/physiology , 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
SELECTION OF CITATIONS
SEARCH DETAIL
...