Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
Add more filters

Publication year range
1.
Cell Commun Signal ; 22(1): 247, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38689280

ABSTRACT

BACKGROUND: Renal fibrosis is a prevalent manifestation of chronic kidney disease (CKD), and effective treatments for this disease are currently lacking. Myofibroblasts, which originate from interstitial fibroblasts, aggregate in the renal interstitium, leading to significant accumulation of extracellular matrix and impairment of renal function. The nonreceptor tyrosine kinase c-Abl (encoded by the Abl1 gene) has been implicated in the development of renal fibrosis. However, the precise role of c-Abl in this process and its involvement in fibroblast-myofibroblast transition (FMT) remain poorly understood. METHODS: To investigate the effect of c-Abl in FMT during renal fibrosis, we investigated the expression of c-Abl in fibrotic renal tissues of patients with CKD and mouse models. We studied the phenotypic changes in fibroblast or myofibroblast-specific c-Abl conditional knockout mice. We explored the potential targets of c-Abl in NRK-49F fibroblasts. RESULTS: In this study, fibrotic mouse and cell models demonstrated that c-Abl deficiency in fibroblasts mitigated fibrosis by suppressing fibroblast activation, fibroblast-myofibroblast transition, and extracellular matrix deposition. Mechanistically, c-Abl maintains the stability of the RACK1 protein, which serves as a scaffold for proteins such as c-Abl and focal adhesion kinase at focal adhesions, driving fibroblast activation and differentiation during renal fibrosis. Moreover, specifically targeting c-Abl deletion in renal myofibroblasts could prove beneficial in established kidney fibrosis by reducing RACK1 expression and diminishing the extent of fibrosis. CONCLUSIONS: Our findings suggest that c-Abl plays a pathogenic role in interstitial fibrosis through the regulation of RACK1 protein stabilization and myofibroblast differentiation, suggesting a promising strategy for the treatment of CKD.


Subject(s)
Fibroblasts , Fibrosis , Myofibroblasts , Proto-Oncogene Proteins c-abl , Receptors for Activated C Kinase , Signal Transduction , Animals , Proto-Oncogene Proteins c-abl/metabolism , Proto-Oncogene Proteins c-abl/genetics , Myofibroblasts/metabolism , Myofibroblasts/pathology , Humans , Mice , Fibroblasts/metabolism , Fibroblasts/pathology , Receptors for Activated C Kinase/genetics , Receptors for Activated C Kinase/metabolism , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Kidney/pathology , Kidney/metabolism , Male , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/genetics , Mice, Knockout , Mice, Inbred C57BL
2.
Brain ; 146(10): 4088-4104, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37071594

ABSTRACT

Nuclear to cytoplasmic mislocalization and aggregation of multiple RNA-binding proteins (RBPs), including FUS, are the main neuropathological features of the majority of cases of amyotrophic lateral sclerosis (ALS) and frontotemporal lobular degeneration (FTLD). In ALS-FUS, these aggregates arise from disease-associated mutations in FUS, whereas in FTLD-FUS, the cytoplasmic inclusions do not contain mutant FUS, suggesting different molecular mechanisms of FUS pathogenesis in FTLD that remain to be investigated. We have previously shown that phosphorylation of the C-terminal Tyr526 of FUS results in increased cytoplasmic retention of FUS due to impaired binding to the nuclear import receptor TNPO1. Inspired by the above notions, in the current study we developed a novel antibody against the C-terminally phosphorylated Tyr526 FUS (FUSp-Y526) that is specifically capable of recognizing phosphorylated cytoplasmic FUS, which is poorly recognized by other commercially available FUS antibodies. Using this FUSp-Y526 antibody, we demonstrated a FUS phosphorylation-specific effect on the cytoplasmic distribution of soluble and insoluble FUSp-Y526 in various cells and confirmed the involvement of the Src kinase family in Tyr526 FUS phosphorylation. In addition, we found that FUSp-Y526 expression pattern correlates with active pSrc/pAbl kinases in specific brain regions of mice, indicating preferential involvement of cAbl in the cytoplasmic mislocalization of FUSp-Y526 in cortical neurons. Finally, the pattern of immunoreactivity of active cAbl kinase and FUSp-Y526 revealed altered cytoplasmic distribution of FUSp-Y526 in cortical neurons of post-mortem frontal cortex tissue from FTLD patients compared with controls. The overlap of FUSp-Y526 and FUS signals was found preferentially in small diffuse inclusions and was absent in mature aggregates, suggesting possible involvement of FUSp-Y526 in the formation of early toxic FUS aggregates in the cytoplasm that are largely undetected by commercially available FUS antibodies. Given the overlapping patterns of cAbl activity and FUSp-Y526 distribution in cortical neurons, and cAbl induced sequestration of FUSp-Y526 into G3BP1 positive granules in stressed cells, we propose that cAbl kinase is actively involved in mediating cytoplasmic mislocalization and promoting toxic aggregation of wild-type FUS in the brains of FTLD patients, as a novel putative underlying mechanism of FTLD-FUS pathophysiology and progression.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Lobar Degeneration , Animals , Humans , Mice , Amyotrophic Lateral Sclerosis/metabolism , DNA Helicases/metabolism , Frontotemporal Lobar Degeneration/pathology , Phosphorylation , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Proto-Oncogene Proteins c-abl
3.
Mol Divers ; 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38273156

ABSTRACT

Parkinson's disease (PD) is the most common movement disorder worldwide. PD is primarily associated with the mutation, overexpression, and phosphorylation of α-synuclein. At the molecular level, the upstream protein c-Abl, a tyrosine kinase, has been shown to regulate α-synuclein activation and expression patterns. This study aimed to identify potential c-Abl inhibitors through in silico approaches. Molecular docking was performed using PyRx software, followed by Prime MM-GBSA studies. BBB permeability and toxicity were predicted using CBligand and ProTox-II, respectively. ADME was assessed using QikProp. Molecular dynamics were carried out using Desmond (Academic version). DFT calculations were performed using the Gaussian 16 suite program. The binding scores of the top hits, norimatinib, DB07326, and entinostat were - 11.8 kcal/mol, - 11.8 kcal/mol, and - 10.8 kcal/mol, respectively. These hits displayed drug-likeness with acceptable ADME properties, except for the standard, nilotinib, which violated Lipinski's rule of five. Similarly, the molecular dynamics showed that the top hits remained stable during the 100 ns simulation. DFT results indicate DB04739 as a potent reactive hit. While based on toxicity prediction, entinostat may be a potential candidate for preclinical and clinical testing in PD. Further studies are warranted to confirm the activity and efficacy of these ligands for PD.

4.
J Biol Chem ; 298(4): 101778, 2022 04.
Article in English | MEDLINE | ID: mdl-35231444

ABSTRACT

Cytoskeletal microtubules (MTs) are nucleated from γ-tubulin ring complexes (γTuRCs) located at MT organizing centers (MTOCs), such as the centrosome. However, the exact regulatory mechanism of γTuRC assembly is not fully understood. Here, we showed that the nonreceptor tyrosine kinase c-Abl was associated with and phosphorylated γ-tubulin, the essential component of the γTuRC, mainly on the Y443 residue by in vivo (immunofluorescence and immunoprecipitation) or in vitro (surface plasmon resonance) detection. We further demonstrated that phosphorylation deficiency significantly impaired γTuRC assembly, centrosome construction, and MT nucleation. c-Abl/Arg deletion and γ-tubulin Y443F mutation resulted in an abnormal morphology and compromised spindle function during mitosis, eventually causing uneven chromosome segregation. Our findings reveal that γTuRC assembly and nucleation function are regulated by Abl kinase-mediated γ-tubulin phosphorylation, revealing a fundamental mechanism that contributes to the maintenance of MT function.


Subject(s)
Microtubule-Organizing Center , Microtubules , Proto-Oncogene Proteins c-abl , Tubulin , Centrosome/metabolism , Microtubule-Organizing Center/metabolism , Microtubules/metabolism , Phosphorylation , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Tubulin/genetics , Tubulin/metabolism
5.
Neurochem Res ; 48(2): 418-434, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36239857

ABSTRACT

c-Abl is a non-receptor tyrosine kinase that promotes intracellular apoptotic signaling in prolonged epileptic seizures. PTZ and pilocarpine-induced continuous epileptic convulsions cause neuronal death and gliosis. C-Abl is linked to oxidative stress, neuronal hyperexcitability, mitochondrial malfunction, and subsequent seizures. We investigated the involvement of c-Abl in epileptogenesis by employing its selective inhibitor Imatinib (1 & 3 mg/kg; i.p.) together with conventional medication valproate (110 mg/kg; i.p.) tends to be effective in decreasing seizures threshold provoked by PTZ for 15 days and pilocarpine for 37 days. Further, Imatinib was effective in preventing epileptic seizures arbitrated oxidative stress injury. Oxidative stress has been linked to excitotoxicity that is considered to pathogenic factor in epileptic brain damage. As ELIZA and biochemical estimations showed the high level of c-Abl as an indicator of neuronal oxidative and apoptosis under chronic PTZ & pilocarpine epileptic seizures marked by decreased antioxidants and elevated levels of caspase-3 that were successfully prevented with Imatinib treatment same as valproate (standard drug). Further, the aberrant c-Abl activation is also linked with neuroinflammation that is also predisposing factor in the development of seizures. Selective inhibition of c-Abl by Imatinib also showed anti-inflammatory activity marked with suppressed levels of NF-kB and pro-inflammatory mediators (TNF-alpha, IL-1ß, and IL-6) suggesting the neuroprotective effect of Imatinib same as valproate (standard drug) in epilepsy. Therefore, the current study provides preclinical evidence of Imatinib as a potential treatment for seizures, as well as an understanding of potential role of c-Ablin epilepsy.


Subject(s)
Epilepsy , Status Epilepticus , Animals , Mice , Anticonvulsants/therapeutic use , Disease Models, Animal , Epilepsy/chemically induced , Epilepsy/drug therapy , Imatinib Mesylate/therapeutic use , Pentylenetetrazole/toxicity , Pilocarpine/toxicity , Seizures/chemically induced , Seizures/drug therapy , Seizures/prevention & control , Status Epilepticus/chemically induced , Valproic Acid/pharmacology
6.
Bioorg Med Chem ; 96: 117532, 2023 12 15.
Article in English | MEDLINE | ID: mdl-38006642

ABSTRACT

Abelson tyrosine kinase (c-Abl) is involved in various biological processes in neurodegenerative diseases and is an attractive target for anti-PD (Parkinson's disease) drug discovery. Based on our previous work, we designed several novel c-Abl inhibitors through a conformational constrained strategy and evaluated their pharmacological activities. Among them, compound A6 exhibited superior inhibitory activity against c-Abl than nilotinib in the homogenous time-resolved fluorescence (HTRF) assay. Furthermore, A6 displayed higher neuroprotective effects against SH-SY5Y cell death induced by MPP+ and lower cytotoxicity than that of nilotinib. Molecular modeling revealed that the 1H-pyrrolo[2,3-B]pyridine ring may contribute to the high affinity of A6 for binding to c-Abl. Collectively, these results suggest that A6 deserves further investigation as a c-Abl inhibitor for neurodegenerative disorders.


Subject(s)
Neuroblastoma , Neuroprotective Agents , Parkinson Disease , Humans , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Neuroprotective Agents/pharmacology , Pyrimidines/pharmacology
7.
Int J Mol Sci ; 24(3)2023 Jan 18.
Article in English | MEDLINE | ID: mdl-36768268

ABSTRACT

Brain-derived neurotrophic factor (BDNF) induces activation of the TrkB receptor and several downstream pathways (MAPK, PI3K, PLC-γ), leading to neuronal survival, growth, and plasticity. It has been well established that TrkB signaling regulation is required for neurite formation and dendritic arborization, but the specific mechanism is not fully understood. The non-receptor tyrosine kinase c-Abl is a possible candidate regulator of this process, as it has been implicated in tyrosine kinase receptors' signaling and trafficking, as well as regulation of neuronal morphogenesis. To assess the role of c-Abl in BDNF-induced dendritic arborization, wild-type and c-Abl-KO neurons were stimulated with BDNF, and diverse strategies were employed to probe the function of c-Abl, including the use of pharmacological inhibitors, an allosteric c-Abl activator, and shRNA to downregulates c-Abl expression. Surprisingly, BDNF promoted c-Abl activation and interaction with TrkB receptors. Furthermore, pharmacological c-Abl inhibition and genetic ablation abolished BDNF-induced dendritic arborization and increased the availability of TrkB in the cell membrane. Interestingly, inhibition or genetic ablation of c-Abl had no effect on the classic TrkB downstream pathways. Together, our results suggest that BDNF/TrkB-dependent c-Abl activation is a novel and essential mechanism in TrkB signaling.


Subject(s)
Brain-Derived Neurotrophic Factor , Neurons , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Neurons/metabolism , Receptor, trkB/genetics , Receptor, trkB/metabolism , Signal Transduction , Proto-Oncogene Proteins c-abl
8.
Glia ; 70(6): 1084-1099, 2022 06.
Article in English | MEDLINE | ID: mdl-35156232

ABSTRACT

Oligodendrocytes (OLs), the myelinating cells in the central nervous system (CNS), are differentiated from OL progenitor cells (OPCs). The proliferation of existing OPCs is indispensable for myelination during CNS development and remyelination in response to demyelination stimulation. The transcription factor Olig2 is required for the specification of OLs and is expressed in the OL lineage. However, the post-translational modification of Olig2 in the proliferation of OPCs is poorly understood. Herein, we identified that c-Abl directly phosphorylates Olig2 mainly at the Tyr137 site, and that Olig2 phosphorylation is essential for OPC proliferation. The expression levels of c-Abl gradually decreased with brain development; moreover, c-Abl was highly expressed in OPCs. OL-specific c-Abl knockout at the developmental stage led to an insufficient proliferation of OPCs, a decreased expression of myelin-related genes, and myelination retardation. Accordingly, a c-Abl-specific kinase inhibitor suppressed OPC proliferation in vitro. Furthermore, we observed that OL-specific c-Abl knockout reduced OPC proliferation and remyelination in a cuprizone model of demyelination. In addition, we found that nilotinib, a clinically used c-Abl inhibitor, decreased the expression of myelin basic protein (Mbp) and motor coordination in mice, indicating a neurological side effect of a long-term administration of the c-Abl inhibitor. Thus, we identified the important role of c-Abl in OLs during developmental myelination and remyelination in a disease model.


Subject(s)
Oligodendrocyte Precursor Cells , Animals , Cell Differentiation/physiology , Cell Proliferation , Mice , Mice, Knockout , Myelin Sheath/metabolism , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Transcription Factor 2/metabolism , Oligodendroglia/metabolism , Phosphorylation
9.
Immunology ; 167(2): 181-196, 2022 10.
Article in English | MEDLINE | ID: mdl-35753034

ABSTRACT

As a nonreceptor tyrosine kinase, Abelson tyrosine kinase (c-Abl) was first studied in chronic myelogenous leukaemia, and its role in lymphocytes has been well characterised. c-Abl is involved in B-cell development and CD19-associated B-cell antigen receptor (BCR) signalling. Although c-Abl regulates different metabolic pathways, the role of c-Abl is still unknown in B-cell metabolism. In this study, B-cell-specific c-Abl knockout (KO) mice (Mb1Cre+/- c-Ablfl/fl ) were used to investigate how c-Abl regulates B-cell metabolism and BCR signalling. We found that the levels of activation positive BCR signalling proximal molecules, phosphorylated spleen tyrosine kinase (pSYK) and phosphorylated Bruton tyrosine kinase (pBTK), were decreased, while the level of key negative regulator, phosphorylated SH2-containing inositol phosphatase 1 (pSHIP1), was increased in Mb1Cre+/- c-Ablfl/fl mice. Furthermore, we found c-Abl deficiency weakened the B-cell spreading, formation of BCR signalosomes, and the polymerisation of actin during BCR activation, and also impaired the differentiation of germinal center (GC) B-cells both in quiescent condition and after immunisation. Moreover, B-cell mitochondrial respiration and the expression of B-cell metabolism-regulating molecules were downregulated in c-Abl deficiency mice. Overall, c-Abl, which involved in actin remodelling and B-cell metabolism, positively regulates BCR signalling and promotes GC differentiation.


Subject(s)
Actins , B-Lymphocytes , Fusion Proteins, bcr-abl , Actins/metabolism , Agammaglobulinaemia Tyrosine Kinase/metabolism , Animals , B-Lymphocytes/metabolism , Cell Differentiation , Fusion Proteins, bcr-abl/metabolism , Mice , Phosphorylation , Receptors, Antigen, B-Cell/metabolism , Syk Kinase/genetics , Syk Kinase/metabolism
10.
FASEB J ; 35(7): e21674, 2021 07.
Article in English | MEDLINE | ID: mdl-34115899

ABSTRACT

Current therapeutic approaches to avoid or reverse bronchoconstriction rely primarily on ß2 adrenoceptor agonists (ß-agonists) that regulate pharmacomechanical coupling/cross bridge cycling in airway smooth muscle (ASM). Targeting actin cytoskeleton polymerization in ASM represents an alternative means to regulate ASM contraction. Herein we report the cooperative effects of targeting these distinct pathways with ß-agonists and inhibitors of the mammalian Abelson tyrosine kinase (Abl1 or c-Abl). The cooperative effect of ß-agonists (isoproterenol) and c-Abl inhibitors (GNF-5, or imatinib) on contractile agonist (methacholine, or histamine) -induced ASM contraction was assessed in cultured human ASM cells (using Fourier Transfer Traction Microscopy), in murine precision cut lung slices, and in vivo (flexiVent in mice). Regulation of intracellular signaling that regulates contraction (pMLC20, pMYPT1, pHSP20), and actin polymerization state (F:G actin ratio) were assessed in cultured primary human ASM cells. In each (cell, tissue, in vivo) model, c-Abl inhibitors and ß-agonist exhibited additive effects in either preventing or reversing ASM contraction. Treatment of contracted ASM cells with c-Abl inhibitors and ß-agonist cooperatively increased actin disassembly as evidenced by a significant reduction in the F:G actin ratio. Mechanistic studies indicated that the inhibition of pharmacomechanical coupling by ß-agonists is near optimal and is not increased by c-Abl inhibitors, and the cooperative effect on ASM relaxation resides in further relaxation of ASM tension development caused by actin cytoskeleton depolymerization, which is regulated by both ß-agonists and c-Abl inhibitors. Thus, targeting actin cytoskeleton polymerization represents an untapped therapeutic reserve for managing airway resistance.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Drug Synergism , Muscle Contraction , Muscle Relaxation , Muscle, Smooth/physiology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Trachea/physiology , Actin Cytoskeleton/metabolism , Animals , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Humans , Imatinib Mesylate/pharmacology , Isoproterenol/pharmacology , Mice , Mice, Inbred C57BL , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Pyrimidines/pharmacology , Signal Transduction , Trachea/cytology , Trachea/drug effects
11.
Bioorg Med Chem ; 74: 117069, 2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36283251

ABSTRACT

C-Abl is involved in various biological processes and plays an important role in neurodegenerative diseases, especially Parkinson's disease (PD). Previous studies have found that nilotinib shows a neuroprotective effect in cell and animal models of PD by inhibiting the activation of c-Abl. But the low blood-brain barrier permeability and potential toxicity limit the further use of nilotinib in PD. Based on molecular modeling studies, a series of 4-methyl-3-(pyridin-2-ylamino)benzamide derivatives were designed and synthesized. In particular, compound 9a exhibited significant inhibitory activity against c-Abl and a potent neuroprotective effect against MPP+-induced SH-SY5Y cell death. Moreover, 9a not only displayed lower cell toxicity compared with nilotinib, but also showed higher oral bioavailability and proper permeability of the blood-brain barrier. This paper provides 4-methyl-3-(pyridin-2-ylamino)benzamide derivatives as a new scaffold for c-Abl inhibitor with potential neuroprotective effect.


Subject(s)
Neuroblastoma , Neuroprotective Agents , Parkinson Disease , Animals , Humans , Neuroprotective Agents/pharmacology , Neuroprotective Agents/metabolism , Neuroblastoma/metabolism , Blood-Brain Barrier/metabolism , Parkinson Disease/metabolism , Benzamides/pharmacology , Benzamides/metabolism , Cell Line, Tumor
12.
Brain ; 144(12): 3674-3691, 2021 12 31.
Article in English | MEDLINE | ID: mdl-34581802

ABSTRACT

Aberrant activation of the non-receptor kinase c-Abl is implicated in the development of pathogenic hallmarks of Parkinson's disease, such as α-synuclein aggregation and progressive neuronal loss. c-Abl-mediated phosphorylation and inhibition of parkin ligase function lead to accumulation of parkin interacting substrate (PARIS) that mediates α-synuclein pathology-initiated dopaminergic neurodegeneration. Here we show that, in addition to PARIS accumulation, c-Abl phosphorylation of PARIS is required for PARIS-induced cytotoxicity. c-Abl-mediated phosphorylation of PARIS at Y137 (within the Krüppel-associated box domain) drives its association with KAP1 and the repression of genes with diverse functions in pathways such as chromatin remodelling and p53-dependent cell death. One phosphorylation-dependent PARIS target, MDM4 (a p53 inhibitor that associates with MDM2; also known as MDMX), is transcriptionally repressed in a histone deacetylase-dependent manner via PARIS binding to insulin response sequence motifs within the MDM4 promoter. Virally induced PARIS transgenic mice develop c-Abl activity-dependent Parkinson's disease features such as motor deficits, dopaminergic neuron loss and neuroinflammation. PARIS expression in the midbrain resulted in c-Abl activation, PARIS phosphorylation, MDM4 repression and p53 activation, all of which are blocked by the c-Abl inhibitor nilotinib. Importantly, we also observed aberrant c-Abl activation and PARIS phosphorylation along with PARIS accumulation in the midbrain of adult parkin knockout mice, implicating c-Abl in recessive Parkinson's disease. Inhibition of c-Abl or PARIS phosphorylation by nilotinib or Y137F-PARIS expression in adult parkin knockout mice blocked MDM4 repression and p53 activation, preventing motor deficits and dopaminergic neurodegeneration. Finally, we found correlative increases in PARIS phosphorylation, MDM4 repression and p53 activation in post-mortem Parkinson's disease brains, pointing to clinical relevance of the c-Abl-PARIS-MDM4-p53 pathway. Taken together, our results describe a novel mechanism of epigenetic regulation of dopaminergic degeneration downstream of pathological c-Abl activation in Parkinson's disease. Since c-Abl activation has been shown in sporadic Parkinson's disease, PARIS phosphorylation might serve as both a useful biomarker and a potential therapeutic target to regulate neuronal loss in Parkinson's disease.


Subject(s)
Dopaminergic Neurons/pathology , Nerve Degeneration/pathology , Parkinsonian Disorders/pathology , Proto-Oncogene Proteins c-abl/metabolism , Repressor Proteins/metabolism , Animals , Dopaminergic Neurons/metabolism , Humans , Mice , Mice, Transgenic , Nerve Degeneration/metabolism , Parkinsonian Disorders/metabolism , Phosphorylation
13.
Int J Mol Sci ; 23(24)2022 Dec 18.
Article in English | MEDLINE | ID: mdl-36555805

ABSTRACT

Endoplasmic reticulum stress activates inositol-requiring enzyme 1α (IRE1α) and protein kinase, R-like endoplasmic reticulum kinase (PERK), the two principal regulators of the unfolded protein response (UPR). In multiple myeloma, adaptive IRE1α signaling is predominantly activated and regulates cell fate along with PERK. Recently, we demonstrated that GNF-2, an allosteric c-Abl inhibitor, rheostatically enhanced IRE1α activity and induced apoptosis through c-Abl conformational changes in pancreatic ß cells. Herein, we analyzed whether the pharmacological modulation of c-Abl conformation resulted in anti-myeloma effects. First, we investigated the effects of GNF-2 on IRE1α activity and cell fate, followed by an investigation of the anti-myeloma effects of asciminib, a new allosteric c-Abl inhibitor. Finally, we performed RNA sequencing to characterize the signaling profiles of asciminib. We observed that both GNF-2 and asciminib decreased cell viability and induced XBP1 mRNA splicing in primary human myeloma cells and myeloma cell lines. RNA sequencing identified the induction of UPR- and apoptosis-related genes by asciminib. Asciminib re-localized c-Abl to the endoplasmic reticulum, and its combination with a specific IRE1α inhibitor, KIRA8, enhanced cell death with the reciprocal induction of CHOP mRNA expression. Together, the allosteric inhibition of c-Abl-activated UPR with anti-myeloma effects; this could be a novel therapeutic target for multiple myeloma.


Subject(s)
Multiple Myeloma , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , Unfolded Protein Response , Endoplasmic Reticulum Stress , Cell Death , RNA, Messenger/genetics , X-Box Binding Protein 1/metabolism
14.
J Cell Sci ; 132(1)2019 01 02.
Article in English | MEDLINE | ID: mdl-30559247

ABSTRACT

The tyrosine kinase c-Abl participates in the regulation of various cellular functions including cell proliferation, adhesion, migration, smooth muscle contraction and cancer progression. However, knowledge regarding transcriptional regulation of c-Abl is surprisingly limited. Sp1 is a founding member of the Sp1 transcription factor family that has been implicated in housekeeping gene expression, tumor cell proliferation and differentiation. Here, we show that knockdown and rescue of Sp1 affected growth factor-mediated c-Abl expression in cells. c-Abl promoter activity was also affected by Sp1 knockdown. This is the first evidence to suggest that Sp1 is an important transcription factor to regulate c-Abl expression. In addition, Sp1 phosphorylation at Thr-453 and Thr-739 has been proposed to regulate its activity in Drosophila cells. We unexpectedly found that growth factors did not induce Sp1 phosphorylation at these two residues. In contrast, growth factor stimulation upregulated Sp1 expression. Intriguingly, inhibition of ERK1 and ERK2 (ERK1/2, also known as MAPK3 and MAPK1, respectively) reduced expression of Sp1 and c-Abl. Furthermore, c-Abl knockdown diminished ERK1/2 phosphorylation and Sp1 expression. Taken together, these studies suggest that Sp1 can modulate c-Abl expression at transcription level. Conversely, c-Abl affects ERK1/2 activation and Sp1 expression in cells.


Subject(s)
Cell Proliferation , Gene Expression Regulation , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Smooth Muscle/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Sp1 Transcription Factor/metabolism , Bronchi/cytology , Bronchi/metabolism , Cells, Cultured , Humans , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Myocytes, Smooth Muscle/cytology , Phosphorylation , Promoter Regions, Genetic , Proto-Oncogene Proteins c-abl/genetics , Signal Transduction , Sp1 Transcription Factor/genetics , Transcriptional Activation
15.
Exp Cell Res ; 388(2): 111808, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31891685

ABSTRACT

Recent studies have shown that serum secretory phospholipase A2 group IB (sPLA2-IB) is associated with proteinuric kidney diseases and plays a pivotal role in podocyte injury via its natural receptor. Arachidonic acid (AA), as a major metabolite of sPLA2-IB, regulates the actin bungling remodeling and contributes to the podocyte injury. However, the underlying mechanism of AA in the regulation of podocyte actin remodeling and human podocyte injury is unclear. Here, we reported that AA induced F-actin cytoskeletal ring formation and promoted protein kinase A (PKA), nephrin and c-Abl phosphorylation. Moreover, AA promoted c-Abl translocation from the nucleus to the cytoplasm and increased the recruitment of c-Abl to p-nephrin by the interaction between them. H89 (PKA inhibitor) provided protection against AA-induced F-actin bunching remodeling, down-regulated nephrin phosphorylation, and suppressed the c-Abl translocation and activation. STI571 (c-Abl inhibitor) also improved the AA associated F-actin bunching remodeling. In addition, H89 and STI571 both alleviated apoptosis and adhesion damage of podocyte. These results indicate that an excess of AA treatment is detrimental to the podocyte actin cytoskeleton and promotes podocyte injury due to the activation of PKA-c-Abl signaling.


Subject(s)
Actin Cytoskeleton/drug effects , Arachidonic Acid/pharmacology , Gene Expression Regulation/drug effects , Podocytes/pathology , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Cells, Cultured , Humans , Podocytes/drug effects , Podocytes/metabolism , Protein Kinase C/genetics , Proto-Oncogene Proteins c-abl/genetics
16.
Mol Pain ; 16: 1744806920930855, 2020.
Article in English | MEDLINE | ID: mdl-32498644

ABSTRACT

Trigeminal neuralgia is a common neuropathic pain in the head and face. The pathogenesis of trigeminal neuralgia is complex, and so far, the pathogenesis of trigeminal neuralgia involving peripheral and central nervous inflammation theory has not been explained clearly. The loss of dopamine neurons in striatum may play an important role in the development of trigeminal nerve, but the reason is not clear. C-Abl is a nonreceptor tyrosine kinase, which can be activated abnormally in the environment of neuroinflammation and cause neuron death. We found that in the rat model of infraorbital nerve ligation trigeminal neuralgia, the pain threshold decreased, the expression of c-Abl increased significantly, the downstream activation product p38 was also activated abnormally and the loss of dopamine neurons in striatum increased. When treated with imatinib mesylate (STI571), a specific c-Abl family kinase inhibitor, the p38 expression was decreased and the loss of dopaminergic neurons was reduced. The mechanical pain threshold of rats was also improved. In conclusion, c-abl-p38 signaling pathway may play an important role in the pathogenesis of trigeminal neuralgia, and it is one of the potential targets for the treatment of trigeminal neuralgia.


Subject(s)
Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Proto-Oncogene Proteins c-abl/metabolism , Signal Transduction , Trigeminal Neuralgia/metabolism , Trigeminal Neuralgia/pathology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Disease Models, Animal , Dopaminergic Neurons/drug effects , Imatinib Mesylate/pharmacology , Male , Models, Biological , Neostriatum/pathology , Nerve Tissue/drug effects , Nerve Tissue/pathology , Pain Threshold/drug effects , Phosphorylation/drug effects , Rats, Sprague-Dawley , Signal Transduction/drug effects
17.
J Recept Signal Transduct Res ; 40(4): 365-373, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32131672

ABSTRACT

Context: Oocyte and granulosa cells (GCs) have bidirectional communication and GCs play an important role in folliculogenesis and proliferation of GCs is very important for the development of ovulatory follicle. DNA double-strand breaks activate c-Abl protein tyrosine kinase and c-Abl has a functional role in repairement of DNA and control of telomere.Objective: In this study, we hypothesized that c-Abl has a regulative role on mTERT in mouse ovarian granulosa cells (GCs) and we aimed to detect c-Abl and mTERT interaction in mouse primary culture of GCs.Materials and methods: Mouse ovarian granulosa cell were cultured and siRNA-mediated knockdown approach was used to knockdown c-Abl expression.Results: We showed c-Abl and mTERT immunolocalization in vivo and in vitro mouse GCs. c-Abl and mTERT were constitutively expressed in mouse granulosa cells and c-Abl presented more intense expression in granulosa cells than mTERT expression. The interaction of the c-Abl-mTERT is supported by the exhibition that c-Abl siRNA knockdown cells show decreased mTERT expression. We also present an interaction between c-Abl and mTERT by immunoprecipitation. In addition, our results indicated that the down-regulation of c-Abl was also accompanied by reduced expression of proliferating cell nuclear antigen (PCNA) in GCs.Conclusions: We suggest that mTERT may associate with the c-Abl in mouse GCs and the interactions between c-Abl and mTERT suggest a role for c-Abl in the regulation of telomerase function and proliferation in mouse granulosa cells.


Subject(s)
Genes, abl/genetics , Granulosa Cells/metabolism , Protein-Tyrosine Kinases/genetics , Telomerase/genetics , Animals , Catalytic Domain/genetics , Cell Proliferation , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation, Developmental/genetics , Granulosa Cells/physiology , Mice , Oocytes/growth & development , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Ovulation/genetics , Protein Interaction Maps/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Telomerase/chemistry
18.
Int J Mol Sci ; 21(19)2020 Oct 01.
Article in English | MEDLINE | ID: mdl-33019757

ABSTRACT

The intermediate filament synemin has been previously identified as novel regulator of cancer cell therapy resistance and DNA double strand break (DSB) repair. c-Abl tyrosine kinase is involved in both of these processes. Using PamGene technology, we performed a broad-spectrum kinase activity profiling in three-dimensionally, extracellular matrix grown head and neck cancer cell cultures. Upon synemin silencing, we identified 86 deactivated tyrosine kinases, including c-Abl, in irradiated HNSCC cells. Upon irradiation and synemin inhibition, c-Abl hyperphosphorylation on tyrosine (Y) 412 and threonine (T) 735 was significantly reduced, prompting us to hypothesize that c-Abl tyrosine kinase is an important signaling component of the synemin-mediated radioresistance pathway. Simultaneous targeting of synemin and c-Abl resulted in similar radiosensitization and DSB repair compared with single synemin depletion, suggesting synemin as an upstream regulator of c-Abl. Immunoprecipitation assays revealed a protein complex formation between synemin and c-Abl pre- and post-irradiation. Upon pharmacological inhibition of ATM, synemin/c-Abl protein-protein interactions were disrupted implying synemin function to depend on ATM kinase activity. Moreover, deletion of the SH2 domain of c-Abl demonstrated a decrease in interaction, indicating the dependency of the protein-protein interaction on this domain. Mechanistically, radiosensitization upon synemin knockdown seems to be associated with an impairment of DNA repair via regulation of non-homologous end joining independent of c-Abl function. Our data generated in more physiological 3D cancer cell culture models suggest c-Abl as further key determinant of radioresistance downstream of synemin.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , DNA Repair , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Intermediate Filament Proteins/genetics , Proto-Oncogene Proteins c-abl/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation/radiation effects , DNA Breaks, Double-Stranded , DNA, Neoplasm/metabolism , Embryo, Nonmammalian , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/radiotherapy , Humans , Intermediate Filament Proteins/antagonists & inhibitors , Intermediate Filament Proteins/metabolism , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-abl/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Radiation Tolerance/genetics , Signal Transduction , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/radiotherapy , X-Rays , Zebrafish
19.
Biochim Biophys Acta Mol Cell Res ; 1865(9): 1173-1186, 2018 09.
Article in English | MEDLINE | ID: mdl-29807053

ABSTRACT

Yin Yang 1 (YY1) is a multifunctional transcription factor that can activate or repress transcription depending on the promotor and/or the co-factors recruited. YY1 is phosphorylated in various signaling pathways and is critical for different biological functions including embryogenesis, apoptosis, proliferation, cell-cycle regulation and tumorigenesis. Here we report that YY1 is a substrate for c-Abl kinase phosphorylation at conserved residue Y254 in the spacer region. Pharmacological inhibition of c-Abl kinase by imatinib, nilotinib and GZD824, knock-down of c-Abl using siRNA, and the use of c-Abl kinase-dead drastically reduces tyrosine phosphorylation of YY1. Both radioactive and non-radioactive in vitro kinase assays, as well as co-immunoprecipitation in different cell lines, show that the target of c-Abl phosphorylation is tyrosine residue 254. c-Abl phosphorylation has little effect on YY1 DNA binding ability or cellular localization in asynchronous cells. However, functional studies reveal that c-Abl mediated phosphorylation of YY1 regulates YY1's transcriptional ability in vivo. In conclusion, we demonstrate the novel role of c-Abl kinase in regulation of YY1's transcriptional activity, linking YY1 regulation with c-Abl tyrosine kinase signaling pathways.


Subject(s)
Oncogene Proteins v-abl/metabolism , Transcription, Genetic , YY1 Transcription Factor/chemistry , YY1 Transcription Factor/metabolism , Benzamides/pharmacology , Conserved Sequence , Gene Knockout Techniques , Gene Silencing , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Imatinib Mesylate/pharmacology , MCF-7 Cells , Oncogene Proteins v-abl/genetics , Phosphorylation , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Tyrosine/chemistry
20.
J Neurosci ; 38(39): 8345-8363, 2018 09 26.
Article in English | MEDLINE | ID: mdl-30082414

ABSTRACT

Eph receptors play pivotal roles in the axon guidance of retinal ganglion cells (RGCs) at the optic chiasm and the establishment of the topographic retinocollicular map. We previously demonstrated that protein tyrosine phosphatase receptor type O (PTPRO) is specifically involved in the control of retinotectal projections in chicks through the dephosphorylation of EphA and EphB receptors. We subsequently revealed that all the mouse R3 subfamily members (PTPRB, PTPRH, PTPRJ, and PTPRO) of the receptor protein tyrosine phosphatase (RPTP) family inhibited Eph receptors as their substrates in cultured mammalian cells. We herein investigated the functional roles of R3 RPTPs in the projection of mouse retinal axon of both sexes. Ptpro and Ptprj were expressed in mouse RGCs; however, Ptprj expression levels were markedly higher than those of Ptpro Consistent with their expression levels, Eph receptor activity was significantly enhanced in Ptprj-knock-out (Ptprj-KO) retinas. In Ptprj-KO and Ptprj/Ptpro-double-KO (DKO) mice, the number of retinal axons that projected ipsilaterally or to the contralateral eye was significantly increased. Furthermore, retinal axons in Ptprj-KO and DKO mice formed anteriorly shifted ectopic terminal zones in the superior colliculus (SC). We found that c-Abl (Abelson tyrosine kinase) was downstream of ephrin-Eph signaling for the repulsion of retinal axons at the optic chiasm and in the SC. c-Abl was identified as a novel substrate for PTPRJ and PTPRO, and the phosphorylation of c-Abl was upregulated in Ptprj-KO and DKO retinas. Thus, PTPRJ regulates retinocollicular projections in mice by controlling the activity of Eph and c-Abl kinases.SIGNIFICANCE STATEMENT Correct retinocollicular projection is a prerequisite for proper vision. Eph receptors have been implicated in retinal axon guidance at the optic chiasm and the establishment of the topographic retinocollicular map. We herein demonstrated that protein tyrosine phosphatase receptor type J (PTPRJ) regulated retinal axonal projections by controlling Eph activities. The retinas of Ptprj-knock-out (KO) and Ptpro/Ptprj double-KO mice exhibited significantly enhanced Eph activities over those in wild-type mice, and their axons showed defects in pathfinding at the chiasm and retinocollicular topographic map formation. We also revealed that c-Abl (Abelson tyrosine kinase) downstream of Eph receptors was regulated by PTPRJ. These results indicate that the regulation of the ephrin-Eph-c-Abl axis by PTPRJ plays pivotal roles in the proper central projection of retinal axons during development.


Subject(s)
Axons/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Receptors, Eph Family/metabolism , Retina/metabolism , Retinal Ganglion Cells/metabolism , Superior Colliculi/metabolism , Animals , Cells, Cultured , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Retina/cytology , Retina/growth & development , Retinal Ganglion Cells/cytology , Superior Colliculi/growth & development , Up-Regulation , Visual Pathways/cytology , Visual Pathways/growth & development , Visual Pathways/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL