Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 129
1.
Int J Mol Sci ; 23(21)2022 Oct 31.
Article En | MEDLINE | ID: mdl-36362028

Bladder cancer is a common global cancer with a high percentage of metastases and high mortality rate. Thus, it is necessary to identify new biomarkers that can be helpful in diagnosis. Pyruvate dehydrogenase kinase 4 (PDK4) belongs to the PDK family and plays an important role in glucose utilization in living organisms. In the present study, we evaluated the role of PDK4 in bladder cancer and its related protein changes. First, we observed elevated PDK4 expression in high-grade bladder cancers. To screen for changes in PDK4-related proteins in bladder cancer, we performed a comparative proteomic analysis using PDK4 knockdown cells. In bladder cancer cell lines, PDK4 silencing resulted in a lower rate of cell migration and invasion. In addition, a PDK4 knockdown xenograft model showed reduced bladder cancer growth in nude mice. Based on our results, PDK4 plays a critical role in the metastasis and growth of bladder cancer cells through changes in ERK, SRC, and JNK.


Protein Kinase Inhibitors , Urinary Bladder Neoplasms , Animals , Humans , Mice , MAP Kinase Signaling System/drug effects , Mice, Nude , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proteomics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , src-Family Kinases/drug effects , src-Family Kinases/metabolism
2.
Fluids Barriers CNS ; 19(1): 7, 2022 Jan 21.
Article En | MEDLINE | ID: mdl-35062973

BACKGROUND: C-C chemokine receptor type 1 (CCR1) and its endogenous ligand, CCL5, participate in the pathogenesis of neuroinflammatory diseases. However, much remains unknown regarding CCL5/CCR1 signaling in blood-brain barrier (BBB) permeability after intracerebral hemorrhage (ICH). METHODS: A total of 250 CD1 male mice were used and ICH was induced via autologous whole blood injection. Either Met-RANTES, a selective CCR1 antagonist, or Met-RANTES combined with a Rac1 CRISPR activator was administered to the mice 1 h after ICH. Post-ICH assessments included neurobehavioral tests, brain water content, BBB integrity, hematoma volume, Western blot, and immunofluorescence staining. The CCR1 ligand, rCCL5, and SRC CRISPR knockout in naïve mice were used to further elucidate detrimental CCL5/CCR1/SRC signaling. RESULTS: Brain endogenous CCR1 and CCL5 were upregulated after ICH in mice with a peak at 24 h, and CCR1 was expressed in endothelial cells, astrocytes, and neurons. Met-R treatment reduced brain edema and neurobehavioral impairment, as well as preserved BBB integrity and tight junction protein expression in ICH mice. Met-R treatment decreased expression of p-SRC, Rac1, albumin, and MMP9, but increased claudin-5, occludin, and ZO-1 tight junction proteins after ICH. These effects were regressed using the Rac1 CRISPR activator. Administration of rCCL5 in naïve mice increased expression of p-SRC, Rac1, albumin, and MMP9, but decreased levels of claudin-5, occludin, and ZO-1 tight junction proteins. These effects in naïve mice were reversed with SRC CRISPR (KO). CONCLUSIONS: Our findings demonstrate that CCR5 inhibition by Met-R improves neurological deficits after ICH by preserving BBB integrity through inhibiting CCR1/SRC/Rac1 signaling pathway in mice. Thus, Met-R has therapeutic potential in the management of ICH patients.


Blood-Brain Barrier/metabolism , CCR5 Receptor Antagonists/pharmacology , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/metabolism , Chemokine CCL5/pharmacology , Neuropeptides/metabolism , Receptors, CCR1/metabolism , rac1 GTP-Binding Protein/metabolism , src-Family Kinases/metabolism , Animals , Blood-Brain Barrier/drug effects , Chemokine CCL5/administration & dosage , Male , Mice , Neuropeptides/drug effects , Receptors, CCR1/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , rac1 GTP-Binding Protein/drug effects , src-Family Kinases/drug effects
3.
Nat Commun ; 12(1): 4099, 2021 07 02.
Article En | MEDLINE | ID: mdl-34215742

The inside of a cell is highly crowded with proteins and other biomolecules. How proteins express their specific functions together with many off-target proteins in crowded cellular environments is largely unknown. Here, we investigate an inhibitor binding with c-Src kinase using atomistic molecular dynamics (MD) simulations in dilute as well as crowded protein solution. The populations of the inhibitor, 4-amino-5-(4-methylphenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP1), in bulk solution and on the surface of c-Src kinase are reduced as the concentration of crowder bovine serum albumins (BSAs) increases. This observation is consistent with the reduced PP1 inhibitor efficacy in experimental c-Src kinase assays in addition with BSAs. The crowded environment changes the major binding pathway of PP1 toward c-Src kinase compared to that in dilute solution. This change is explained based on the population shift mechanism of local conformations near the inhibitor binding site in c-Src kinase.


Protein Kinase Inhibitors/pharmacology , Proteins/metabolism , src-Family Kinases/drug effects , src-Family Kinases/metabolism , Animals , Binding Sites , CSK Tyrosine-Protein Kinase/drug effects , CSK Tyrosine-Protein Kinase/metabolism , Computational Biology , Models, Molecular , Proteins/chemistry , Pyrazoles/pharmacology , Pyrimidines/pharmacology , src-Family Kinases/chemistry
4.
Biochim Biophys Acta Mol Cell Res ; 1866(9): 1412-1420, 2019 09.
Article En | MEDLINE | ID: mdl-31176697

Src is a known proto-oncogene and its aberrant activity is involved in a variety of cancers, including ovarian cancer, whereas the regulatory mechanism of Src has not been fully clarified. In this study, we identified tripartite motif-containing (TRIM) 50 as a novel negative regulator of Src protein. Our data showed that TRIM50 directly interacted with SH3 domain of Src via its B-box domain; and TRIM50 reduced Src stability by inducing RING domain-dependent K48-linked poly-ubiquitous modification. We further demonstrated that TRIM50 acted as a tumor suppressor in ovarian cancer cells by its negative regulation of Src protein. In vivo animal model verified that TRIM50 inhibited the xenograft tumor growth of ovarian cancer by suppressing Src protein. Clinical investigation showed that expression of TRIM50 in clinical specimens was inversely correlated with the clinical stages, pathology grades and lymph node metastatic status of the patients, which indicated the involvement of aberrant TRIM50 expression in disease progression. Further analysis verified the negative correlation between TRIM50 and Src expression in clinical specimens. Altogether, we identified TRIM50 as a novel suppressor of Src protein, and demonstrated that TRIM50 inhibited ovarian cancer progression by targeting Src and reducing its activity, which provided a novel therapeutic strategy for Src over-activated cancers by positive regulation of TRIM50.


Disease Progression , Ovarian Neoplasms/drug therapy , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/pharmacology , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/pharmacology , src-Family Kinases/drug effects , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Models, Animal , Female , HEK293 Cells , Humans , Lymph Nodes , Middle Aged , Ovarian Neoplasms/pathology , Proto-Oncogene Mas , Xenograft Model Antitumor Assays , src-Family Kinases/metabolism
5.
Life Sci ; 213: 248-257, 2018 Nov 15.
Article En | MEDLINE | ID: mdl-30292831

PURPOSE: Statins extended their hypocholestremic effect to show a promising anticancer activity. Hepatocellular carcinoma (HCC), the third common cause of cancer-related death, responded positively to statins. Some in-vitro studies reveal the rosuvastatin antitumor effect, but barely in-vivo studies. Hence, we evaluated the antitumor potential of rosuvastatin in a HCC model, the possible signaling cues involved, and whether it augments the dasatinib anticancer effect. METHOD: For the in-vitro study, the IC50 and the combination (CI)/dose reduction (DRI) indices were determined for HCC cell line (HepG2) treated with dasatinib and/or rosuvastatin. For the in-vivo study, mice with diethylnitrosamine-induced HCC were treated for 21 days with dasatinib and/or rosuvastatin (10 and 20 mg/kg, respectively). The p-focal adhesion kinase/p-rous sarcoma oncogene cellular homolog (p-FAK/p-Src) cascade and its downstream molecules were assessed. RESULTS: The in-vitro study confirmed the synergistic effect of rosuvastatin with dasatinib, which entailed the in-vivo results. The two drugs decreased the p-FAK/p-Src cue along with p-Ras/c-Raf, p-STAT-3, and p-Akt levels to enhance apoptosis by an increase in caspase-3 level and a decline in survivin level. Additionally, they inhibited HGF, VEGF, and the MMP-9. Moreover, the different treatments downregulated the expression of proliferative cell nuclear antigen (PCNA) and Ki-67. The best effect was mediated by the combination regimen that surpassed the effect of either drug alone. CONCLUSION: Our results highlighted some of the signals involved in rosuvastatin antitumor effect and nominate it as an adds-on therapy with dasatinib to yield a better effect in HCC through inhibiting the FAK/Src cascade.


Dasatinib/pharmacology , Rosuvastatin Calcium/pharmacology , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Focal Adhesion Kinase 1/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Male , Mice , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , src-Family Kinases/drug effects
6.
Cell Adh Migr ; 12(3): 286-298, 2018 05 04.
Article En | MEDLINE | ID: mdl-28678601

Interaction of leukemia blasts with the bone marrow extracellular matrix often results in protection of leukemia cells from chemotherapy and in persistence of the residual disease which is on the basis of subsequent relapses. The adhesion signaling pathways have been extensively studied in adherent cells as well as in mature haematopoietic cells, but the adhesion structures and signaling in haematopoietic stem and progenitor cells, either normal or malignant, are much less explored. We analyzed the interaction of leukemia cells with fibronectin (FN) using interference reflection microscopy, immunofluorescence, measurement of adherent cell fraction, real-time microimpedance measurement and live cell imaging. We found that leukemia cells form very dynamic adhesion structures similar to early stages of focal adhesions. In contrast to adherent cells, where Src family kinases (SFK) belong to important regulators of focal adhesion dynamics, we observed only minor effects of SFK inhibitor dasatinib on leukemia cell binding to FN. The relatively weak involvement of SFK in adhesion structure regulation might be associated with the lack of cytoskeletal mechanical tension in leukemia cells. On the other hand, active Lyn kinase was found to specifically localize to leukemia cell adhesion structures and a less firm cell attachment to FN was often associated with higher Lyn activity (this unexpectedly occurred also after cell treatment with the inhibitor SKI-1). Lyn thus may be important for signaling from integrin-associated complexes to other processes in leukemia cells.


Dasatinib/pharmacology , Fibronectins/metabolism , Leukemia/drug therapy , src-Family Kinases/drug effects , Cell Adhesion/drug effects , Cell Adhesion/physiology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/drug effects , Focal Adhesions/metabolism , Humans , Phosphorylation/drug effects , src-Family Kinases/metabolism
7.
Article En | MEDLINE | ID: mdl-28879169

Herpes simplex virus type 1 (HSV-1) is a ubiquitous pathogen that establishes a latent persistent neuronal infection in humans. The pathogenic effects of repeated viral reactivation in infected neurons are still unknown. Several studies have reported that during HSV-1 epithelial infection, the virus could modulate diverse cell signaling pathways remodeling the Golgi apparatus (GA) membranes, but the molecular mechanisms implicated, and the functional consequences to neurons is currently unknown. Here we report that infection of primary neuronal cultures with HSV-1 triggers Src tyrosine kinase activation and subsequent phosphorylation of Dynamin 2 GTPase, two players with a role in GA integrity maintenance. Immunofluorescence analyses showed that HSV-1 productive neuronal infection caused a scattered and fragmented distribution of the GA through the cytoplasm, contrasting with the uniform perinuclear distribution pattern observed in control cells. In addition, transmission electron microscopy revealed swollen cisternae and disorganized stacks in HSV-1 infected neurons compared to control cells. Interestingly, PP2, a selective inhibitor for Src-family kinases markedly reduced these morphological alterations of the GA induced by HSV-1 infection strongly supporting the possible involvement of Src tyrosine kinase. Finally, we showed that HSV-1 tegument protein VP11/12 is necessary but not sufficient to induce Dyn2 phosphorylation. Altogether, these results show that HSV-1 neuronal infection triggers activation of Src tyrosine kinase, phosphorylation of Dynamin 2 GTPase, and perturbation of GA integrity. These findings suggest a possible neuropathogenic mechanism triggered by HSV-1 infection, which could involve dysfunction of the secretory system in neurons and central nervous system.


GTP Phosphohydrolases/metabolism , Golgi Apparatus/metabolism , Golgi Apparatus/virology , Herpesvirus 1, Human/pathogenicity , src-Family Kinases/metabolism , Animals , Antigens, Viral/metabolism , Cell Line , Cell Membrane/metabolism , Cell Survival , Central Nervous System/metabolism , Central Nervous System/virology , Chlorocebus aethiops , Cytoplasm/metabolism , Cytoplasm/virology , Dynamin II , Dynamins/metabolism , Gene Expression Regulation, Viral , Genes, Viral/genetics , Golgi Apparatus/ultrastructure , Herpesvirus 1, Human/genetics , Humans , Mice , Microscopy, Electron, Transmission , Neurons/metabolism , Neurons/virology , Phosphorylation , Pyrimidines/pharmacology , Signal Transduction , Vero Cells , Viral Proteins/metabolism , src-Family Kinases/drug effects
8.
Arthritis Rheumatol ; 68(1): 117-26, 2016 Jan.
Article En | MEDLINE | ID: mdl-26314228

OBJECTIVE: Oxidative posttranslational modifications of intracellular proteins can potentially regulate signaling pathways relevant to cartilage destruction in arthritis. In this study, oxidation of cysteine residues to form sulfenic acid (S-sulfenylation) was examined in osteoarthritic (OA) chondrocytes and investigated in normal chondrocytes as a mechanism by which fragments of fibronectin (FN-f) stimulate chondrocyte catabolic signaling. METHODS: Chondrocytes isolated from OA and normal human articular cartilage were analyzed using analogs of dimedone that specifically and irreversibly react with protein S-sulfenylated cysteines. Global S-sulfenylation was measured in cell lysates with and without FN-f stimulation by immunoblotting and in fixed cells by confocal microscopy. S-sulfenylation in specific proteins was identified by mass spectroscopy and confirmed by immunoblotting. Src activity was measured in live cells using a fluorescence resonance energy transfer biosensor. RESULTS: Proteins in chondrocytes isolated from OA cartilage were found to have elevated basal levels of S-sulfenylation relative to those of chondrocytes from normal cartilage. Treatment of normal chondrocytes with FN-f induced increased levels of S-sulfenylation in multiple proteins, including the tyrosine kinase Src. FN-f treatment also increased the levels of Src activity. Pretreatment with dimedone to alter S-sulfenylation function or with Src kinase inhibitors inhibited FN-f-induced production of matrix metalloproteinase 13. CONCLUSION: These results demonstrate for the first time the presence of oxidative posttranslational modification of proteins in human articular chondrocytes by S-sulfenylation. Due to the ability to regulate the activity of a number of cell signaling pathways, including catabolic mediators induced by fibronectin fragments, S-sulfenylation may contribute to cartilage destruction in OA and warrants further investigation.


Cartilage, Articular/cytology , Chondrocytes/metabolism , Cysteine/metabolism , Osteoarthritis/metabolism , Oxidation-Reduction , Sulfenic Acids/metabolism , src-Family Kinases/metabolism , Adult , Aged , Aged, 80 and over , Case-Control Studies , Chondrocytes/drug effects , Cyclohexanones/pharmacology , Female , Fibronectins/pharmacology , History, Ancient , Humans , Immunoblotting , Mass Spectrometry , Matrix Metalloproteinase 13/drug effects , Matrix Metalloproteinase 13/metabolism , Microscopy, Confocal , Middle Aged , Peptide Fragments/pharmacology , Protein Processing, Post-Translational , Signal Transduction , src-Family Kinases/drug effects
9.
Arthritis Rheumatol ; 68(2): 359-69, 2016 Feb.
Article En | MEDLINE | ID: mdl-26414708

OBJECTIVE: During rheumatoid arthritis (RA), fibroblast-like synoviocytes (FLS) critically promote disease pathogenesis by aggressively invading the extracellular matrix of the joint. The focal adhesion kinase (FAK) signaling pathway is emerging as a contributor to the anomalous behavior of RA FLS. The receptor protein tyrosine phosphatase α (RPTPα), which is encoded by the PTPRA gene, is a key promoter of FAK signaling. The aim of this study was to investigate whether RPTPα mediates FLS aggressiveness and RA pathogenesis. METHODS: Through RPTPα knockdown, we assessed FLS gene expression by quantitative polymerase chain reaction analysis and enzyme-linked immunosorbent assay, invasion and migration by Transwell assays, survival by annexin V and propidium iodide staining, adhesion and spreading by immunofluorescence microscopy, and activation of signaling pathways by Western blotting of FLS lysates. Arthritis development was examined in RPTPα-knockout (KO) mice using the K/BxN serum-transfer model. The contribution of radiosensitive and radioresistant cells to disease was evaluated by reciprocal bone marrow transplantation. RESULTS: RPTPα was enriched in the RA synovial lining. RPTPα knockdown impaired RA FLS survival, spreading, migration, invasiveness, and responsiveness to platelet-derived growth factor, tumor necrosis factor, and interleukin-1 stimulation. These phenotypes correlated with increased phosphorylation of Src on inhibitory Y(527) and decreased phosphorylation of FAK on stimulatory Y(397) . Treatment of RA FLS with an inhibitor of FAK phenocopied the knockdown of RPTPα. RPTPα-KO mice were protected from arthritis development, which was due to radioresistant cells. CONCLUSION: By regulating the phosphorylation of Src and FAK, RPTPα mediates proinflammatory and proinvasive signaling in RA FLS, correlating with the promotion of disease in an FLS-dependent model of RA.


Arthritis, Experimental/genetics , Arthritis, Rheumatoid/genetics , Fibroblasts/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , src-Family Kinases/metabolism , Animals , Ankle Joint , Apoptosis/drug effects , Apoptosis/genetics , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Blotting, Western , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Movement/drug effects , Cell Movement/genetics , Cell Survival/drug effects , Cell Survival/genetics , Disease Progression , Enzyme-Linked Immunosorbent Assay , Fibroblasts/drug effects , Gene Expression Profiling , Gene Knockdown Techniques , Interleukin-1/pharmacology , Mice , Mice, Knockout , Phosphorylation/drug effects , Phosphorylation/genetics , Platelet-Derived Growth Factor/pharmacology , Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Synovial Membrane/cytology , Tumor Necrosis Factor-alpha/pharmacology , src-Family Kinases/drug effects
10.
Am J Physiol Endocrinol Metab ; 310(3): E225-37, 2016 Feb 01.
Article En | MEDLINE | ID: mdl-26646098

Salidroside (SAL) is a phenylethanoid glycoside isolated from the medicinal plant Rhodiola rosea. R. rosea has been reported to have beneficial effects on diabetic nephropathy (DN) and high-glucose (HG)-induced mesangial cell proliferation. Given the importance of caveolin-1 (Cav-1) in transcytosis of albumin across the endothelial barrier, the present study was designed to elucidate whether SAL could inhibit Cav-1 phosphorylation and reduce the albumin transcytosis across glomerular endothelial cells (GECs) to alleviate diabetic albuminuria as well as to explore its upstream signaling pathway. To assess the therapeutic potential of SAL and the mechanisms involved in DN albuminuria, we orally administered SAL to db/db mice, and the effect of SAL on the albuminuria was measured. The albumin transcytosis across GECs was explored in a newly established in vitro cellular model. The ratio of albumin to creatinine was significantly reduced upon SAL treatment in db/db mice. SAL decreased the albumin transcytosis across GECs in both normoglycemic and hyperglycemic conditions. SAL reversed the HG-induced downregulation of AMP-activated protein kinase and upregulation of Src kinase and blocked the upregulation Cav-1 phosphorylation. Meanwhile, SAL decreased mitochondrial superoxide anion production and moderately depolarized mitochondrial membrane potential. We conclude that SAL exerts its proteinuria-alleviating effects by downregulation of Cav-1 phosphorylation and inhibition of albumin transcytosis across GECs. These studies provide the first evidence of interference with albumin transcytosis across GECs as a novel approach to the treatment of diabetic albuminuria.


Albumins/drug effects , Albuminuria/metabolism , Caveolin 1/drug effects , Diabetic Nephropathies/metabolism , Endothelial Cells/drug effects , Glucosides/pharmacology , Kidney Glomerulus/drug effects , Phenols/pharmacology , Transcytosis/drug effects , AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Albumins/metabolism , Animals , Caveolin 1/metabolism , Creatinine/metabolism , Endothelial Cells/metabolism , Glucose/metabolism , In Vitro Techniques , Kidney Glomerulus/metabolism , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Mice , Phosphorylation/drug effects , src-Family Kinases/drug effects , src-Family Kinases/metabolism
11.
Life Sci ; 144: 208-17, 2016 Jan 01.
Article En | MEDLINE | ID: mdl-26656263

AIMS: Hydrogen sulfide (H2S) ameliorates cardiac fibrosis in several models by suppressing endoplasmic reticulum (ER) stress. Endothelial-to-mesenchymal transition (EndMT) is implicated in the development of cardiac fibrosis. Therefore, we investigated whether H2S could attenuate EndMT by suppressing ER stress. MAIN METHODS: ER stress was induced by tunicamycin (TM) and thapsigargin (TG) and inhibited by 4-phenylbutyrate (4-PBA) in human umbilical vein endothelial cells (HUVECs). ER stress and EndMT were measured by Western blot, Real-Time PCR and immunofluorescence staining. Inhibition Smad2 and Src pathway were performed by specific inhibitors and siRNA. Ultrastructural examination was detected by transmission electron microscope. The functions of HUVECs were investigated by cell migration assay and tube formation in vitro. KEY FINDINGS: Under ER stress, the expression of endothelial marker CD31 significantly decreased while mesenchymal markers α-SMA, vimentin and collagen 1 increased which could be inhibited by 4-PBA. Moreover, HUVECs changed into a fibroblast-like appearance with the activation of Smad2 and Src kinase pathway. After inhibiting Src pathway, EndMT would be significantly inhibited. TM reduced H2S levels in cell lysate and H2S pretreatment could preserve endothelial cell appearance with decreased ER stress and ameliorated dilation of ER. H2S could also downregulate the mesenchymal marker expression, and upregulate the endothelial markers expression, accompanied with the suppression of Src pathway. Moreover, H2S partially restored the capacity of migration and tube formation in HUVECs. SIGNIFICANCE: These results revealed that H2S could protect against ER stress-induced EndMT through Src pathway, which may be a novel role for the cardioprotection of H2S.


Endoplasmic Reticulum Stress/drug effects , Epithelial-Mesenchymal Transition/drug effects , Hydrogen Sulfide/pharmacology , src-Family Kinases/drug effects , src-Family Kinases/physiology , Down-Regulation/drug effects , Fibrosis , Human Umbilical Vein Endothelial Cells , Humans , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Smad2 Protein/antagonists & inhibitors , Smad2 Protein/physiology , Unfolded Protein Response/drug effects
12.
Pain ; 156(12): 2572-2584, 2015 Dec.
Article En | MEDLINE | ID: mdl-26407042

Treating neuropathic pain continues to be a major clinical challenge and underlying mechanisms of neuropathic pain remain elusive. We have recently demonstrated that Wnt signaling, which is important in developmental processes of the nervous systems, plays critical roles in the development of neuropathic pain through the ß-catenin-dependent pathway in the spinal cord and the ß-catenin-independent pathway in primary sensory neurons after nerve injury. Here, we report that Wnt signaling may contribute to neuropathic pain through the atypical Wnt/Ryk signaling pathway in rats. Sciatic nerve injury causes a rapid-onset and long-lasting expression of Wnt3a, Wnt5b, and Ryk receptors in primary sensory neurons, and dorsal horn neurons and astrocytes. Spinal blocking of the Wnt/Ryk receptor signaling inhibits the induction and persistence of neuropathic pain without affecting normal pain sensitivity and locomotor activity. Blocking activation of the Ryk receptor with anti-Ryk antibody, in vivo or in vitro, greatly suppresses nerve injury-induced increased intracellular Ca and hyperexcitability of the sensory neurons, and also the enhanced plasticity of synapses between afferent C-fibers and the dorsal horn neurons, and activation of the NR2B receptor and the subsequent Ca-dependent signals CaMKII, Src, ERK, PKCγ, and CREB in sensory neurons and the spinal cord. These findings indicate a critical mechanism underlying the pathogenesis of neuropathic pain and suggest that targeting the Wnt/Ryk signaling may be an effective approach for treating neuropathic pain.


Astrocytes/metabolism , Behavior, Animal/physiology , Neuralgia/metabolism , Neuronal Plasticity/physiology , Posterior Horn Cells/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Sensory Receptor Cells/metabolism , Wnt Signaling Pathway/physiology , Animals , Behavior, Animal/drug effects , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cyclic AMP Response Element-Binding Protein/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Ganglia, Spinal/metabolism , Hyperalgesia/genetics , Hyperalgesia/metabolism , Locomotion/drug effects , MAP Kinase Signaling System/drug effects , Male , Nerve Fibers, Unmyelinated/drug effects , Neuralgia/genetics , Neuronal Plasticity/drug effects , Pain Threshold/drug effects , Patch-Clamp Techniques , Protein Kinase C , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Sciatic Nerve/injuries , Signal Transduction , Spinal Cord/metabolism , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/metabolism , Wnt Signaling Pathway/drug effects , Wnt3A Protein/antagonists & inhibitors , Wnt3A Protein/metabolism , src-Family Kinases/drug effects , src-Family Kinases/metabolism
13.
Int Immunopharmacol ; 23(2): 434-41, 2014 Dec.
Article En | MEDLINE | ID: mdl-25261409

Epigallocatechin-3-gallate (EGCG) exerts anti-inflammatory properties on immune cells and binds to CD4 molecules. However, the effects of EGCG on CD4(+) T cells remain largely unknown. Here, we found that EGCG enhanced IFN-γ-induced signal transducer and activator of transcription 1 (STAT1) activation in primary CD4(+) T cells from C57BL/6 mice and in a human leukemic CD4(+) T-cell line of Hut 78 cells, while it inhibited the classical pathway of IFN-γ signaling including activating phosphorylations of Janus kinase (JAK) 1, JAK2 and STAT3, forming interferon-γ activated sequence (GAS)-binding STAT1 homodimers, and producing pro-inflammatory chemokine (C-X-C motif) ligand 9 (CXCL9). CD4 blockade did not suppress the increase in IFN-γ-induced STAT1 activation in CD4(+) T cells by EGCG. Furthermore, activation of Src kinase was also triggered by IFN-γ plus EGCG in both Hut 78 and primary CD4(+) T cells. Interestingly, EGCG promoted apoptosis of CD4(+) T cells treated with IFN-γ. The increases in STAT1 activation and apoptosis induced by EGCG in IFN-γ-activated CD4(+) T cells were almost completely abolished by a selective Src family kinase inhibitor, SU6656. Moreover, EGCG alleviates CD4(+) CD45RB(hi) CD25(-) T cell transfer induced colitis with less accumulation of CD4(+) T cells in the colon. In conclusion, the present study reports an alternative activation of STAT1 via Src by EGCG in IFN-γ-activated CD4(+) T cells, which promotes the apoptosis of IFN-γ-activated CD4(+) T cells and contributes to the improvement of T cell-mediated colitis. Our findings suggest a novel role of EGCG in regulating IFN-γ signaling and controlling inflammation.


Apoptosis/drug effects , CD4-Positive T-Lymphocytes/drug effects , Catechin/analogs & derivatives , Interferon-gamma/pharmacology , STAT1 Transcription Factor/metabolism , Animals , Apoptosis/physiology , CD4-Positive T-Lymphocytes/physiology , Catechin/pharmacology , Cell Line , Female , Humans , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , STAT1 Transcription Factor/genetics , Signal Transduction , Spleen/cytology , src-Family Kinases/drug effects , src-Family Kinases/metabolism , Interferon gamma Receptor
14.
Pharmazie ; 69(5): 346-52, 2014 May.
Article En | MEDLINE | ID: mdl-24855825

Chronic myelogenous leukemia (CML) is triggered by the constitutively activated BCR-ABL oncoprotein and multiple downstream signaling pathways, including the Raf/MEK/ERK, Akt/mTOR, SRC, and STAT5 pathways. The BCR-ABL tyrosine kinase inhibitor imatinib is the standard treatment for CML. However, the development of imatinib resistance has become a new challenge for CML treatment. Here, we investigated the expression levels of the signaling pathways to explore the cause of imatinib resistance and seek new reversing drugs. Our results showed that abnormal activation of the BCR-ABL-independent Lyn/ERK signaling pathway was involved in imatinib-resistance of K562R cells. Furthermore, p-Lyn and p-ERK were up-regulated after treatment with imatinib alone. However, U0126, a MEK1/2 inhibitor, could counteract the up-regulation induced by imatinib, and the combination of imatinib and U0126 could overcome the resistance to imatinib in K562R cells. In conclusion, our studies suggest that the combination of imatinib and an inhibitor of the ERK signaling pathway may be effective in imatinib-resistant CML patients.


Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Butadienes/pharmacology , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Nitriles/pharmacology , Piperazines/pharmacology , Pyrimidines/pharmacology , src-Family Kinases/drug effects , Blotting, Western , Down-Regulation/drug effects , Flow Cytometry , Humans , Imatinib Mesylate , Indicators and Reagents , K562 Cells , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-raf/drug effects , Real-Time Polymerase Chain Reaction , STAT5 Transcription Factor/drug effects , TOR Serine-Threonine Kinases/drug effects , Tetrazolium Salts , Thiazoles
15.
Photodiagnosis Photodyn Ther ; 11(1): 27-33, 2014 Mar.
Article En | MEDLINE | ID: mdl-24280438

BACKGROUND: There are proteins, responsible for many basic cell functions (transmission of extracellular signals to cytoplasm or nucleus, cell growth, proliferation, migration, survival), which are activated and overexpressed in response to acute oxidative stress, especially tyrosine kinases. The oxidative stress-associated Src activator/Homo sapiens chromosome 9 open reading frame 10 protein (Ossa/C9orf10) protects cancer cells from oxidative stress-induced apoptosis by Src family kinases activation. METHODS: In this study precursor of protoporphyrin IX, 5-aminolevulinic acid and its encapsulated form were used in treating MCF-7 human breast cancer cells. After light illumination, cells were collected at different time points and used for evaluation (immunocytochemistry, Western blot analysis) of expression of above proteins, c-Src and Ossa. RESULTS: Our results showed that 5-aminolevulinic acid-mediated photodynamic therapy caused decrease of c-Src expression at 7h after irradiation. The strongest expression was observed at 24h after treatment. Encapsulated form of 5-aminolevulinic acid in terms of PDT caused similar changes of expression of c-Src protein. Furthermore, we observed strong Ossa expression at 7h after treatment in comparison to very low expression at time points 0, 18 and 24h. CONCLUSION: We would like to emphasize that our results showed high expression of Ossa at early time interval after PDT, which was accompanied by a low expression of c-Src kinase, what could protect cancer cells from PDT through activation of c-Src in response to oxidative stress.


Aminolevulinic Acid/pharmacology , Chromosomes, Human, Pair 9/drug effects , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , RNA-Binding Proteins/drug effects , Apoptosis , Cell Line, Tumor , Cell Proliferation , Female , Humans , Immunohistochemistry , MCF-7 Cells , Open Reading Frames , src-Family Kinases/drug effects
16.
Cell Biol Toxicol ; 29(4): 303-19, 2013 Aug.
Article En | MEDLINE | ID: mdl-23955088

Benzo-[a]-pyrene (B[a]P) is a family member of polycyclic aromatic hydrocarbons and a widespread environmental pollutant. It is a mammary carcinogen in rodents and contributes to the development of human breast cancer. However, the signal transduction pathways induced by B[a]P and its role in breast cancer progression have not been studied in detail. Here, we demonstrate that B[a]P induces cell migration through a lipoxygenase- and Src-dependent pathway, as well as the activation of focal adhesion kinase, Src, and the extracellular signal-regulated kinase 2 in MDA-MB-231 breast cancer cells. However, B[a]P is not able to promote migration in the mammary nontumorigenic epithelial cells MCF12A. Moreover, B[a]P promotes an increase of αvß3 integrin-cell surface levels and an increase of metalloproteinase (MMP)-2 and MMP-9 secretions. In summary, our findings demonstrate that B[a]P induces the activation of signal transduction pathways and biological processes involved in the invasion/metastasis process in MDA-MB-231 breast cancer cells.


Benzopyrenes/pharmacology , Cell Movement/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , src-Family Kinases/metabolism , Breast Neoplasms/drug therapy , Cell Line, Tumor , Enzyme Activation/drug effects , Female , Focal Adhesion Protein-Tyrosine Kinases/biosynthesis , Focal Adhesion Protein-Tyrosine Kinases/drug effects , Humans , Integrin alphaVbeta3/biosynthesis , Lipoxygenase/drug effects , Lipoxygenase/metabolism , MCF-7 Cells , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/metabolism , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 1/drug effects , Neoplasm Invasiveness , Neoplasm Metastasis , Signal Transduction/drug effects , src-Family Kinases/biosynthesis , src-Family Kinases/drug effects
17.
Hepatology ; 57(5): 1838-46, 2013 May.
Article En | MEDLINE | ID: mdl-23299860

UNLABELLED: Hepatocellular carcinoma (HCC) is the fifth most common malignancy and is the third leading cause of cancer death worldwide. Recently, the multitargeted kinase inhibitor sorafenib was shown to be the first systemic agent to improve survival in advanced HCC. Unlike other malignancies such as breast cancer, in which molecular subtypes have been clearly defined (i.e., luminal, HER2 amplified, basal, etc.) and tied to effective molecular therapeutics (hormone blockade and trastuzumab, respectively), in HCC this translational link does not exist. Molecular profiling studies of human HCC have identified unique molecular subtypes of the disease. We hypothesized that a panel of human HCC cell lines would maintain molecular characteristics of the clinical disease and could then be used as a model for novel therapeutics. Twenty human HCC cell lines were collected and RNA was analyzed using the Agilent microarray platform. Profiles from the cell lines in vitro recapitulate previously described subgroups from clinical material. Next, we evaluated whether molecular subgroup would have predictive value for response to the Src/Abl inhibitor dasatinib. The results demonstrate that sensitivity to dasatinib was associated with a progenitor subtype. Dasatinib was effective at inducing cell cycle arrest and apoptosis in "progenitor-like" cell lines but not in resistant lines. CONCLUSION: These findings suggest that cell line models maintain the molecular background of HCC and that subtype may be important for selecting patients for response to novel therapies. In addition, it highlights a potential role for Src family signaling in this progenitor subtype of HCC.


Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Thiazoles/therapeutic use , src-Family Kinases/antagonists & inhibitors , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dasatinib , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , In Vitro Techniques , Male , Pharmacogenetics , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering/pharmacology , Thiazoles/pharmacology , src-Family Kinases/drug effects , src-Family Kinases/genetics
18.
Neurobiol Aging ; 34(3): 745-56, 2013 Mar.
Article En | MEDLINE | ID: mdl-22892311

The activity of protein phosphatase (PP) 2A is downregulated and promotes the hyperphosphorylation of tau in the brains of Alzheimer's disease (AD), but the mechanism for PP2A inactivation has not been elucidated. We have reported that PP2A phosphorylation at tyrosine 307 (Y307) is involved in PP2A inactivation. Here, we further studied the upstream mechanisms for PP2A phosphorylation and inactivation. We found that zinc, a heavy metal ion that is widely distributed in the normal brain and accumulated in the susceptible regions of AD brain, could induce PP2A inhibition, phosphorylation of PP2A at Y307 and tau hyperphosphorylation both in rat brains and cultured N2a cells, while zinc chelating prevented these changes completely. Upregulation of PP2A chemically or genetically attenuated zinc-induced tau hyperphosphorylation, whereas mutation of Y307 to phenylalanine abolished the zinc-induced tyrosine phosphorylation and inactivation of PP2A. Zinc could activate Src, while PP2, a specific Src family kinases inhibitor, attenuated zinc-induced PP2A phosphorylation and inactivation, indicating that zinc induces PP2A Y307 phosphorylation and inactivation through Src activation. In human tau transgenic mice, zinc chelator rescued PP2A activity, prevented Src activation, and reduced hyperphosphorylated and insoluble tau levels. We concluded that zinc induces PP2A inactivation and tau hyperphosphorylation through Src-dependent pathway, regulation of zinc homeostasis may be a promising therapeutic for AD and the related tauopathies.


Brain , Protein Phosphatase 2 , Tauopathies/metabolism , Trace Elements/pharmacology , Zinc/pharmacology , src-Family Kinases , tau Proteins , Animals , Brain/drug effects , Brain/metabolism , Cell Line, Tumor , Humans , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Phosphatase 2/drug effects , Protein Phosphatase 2/metabolism , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Tyrosine/drug effects , Tyrosine/metabolism , src-Family Kinases/drug effects , src-Family Kinases/metabolism , tau Proteins/drug effects , tau Proteins/metabolism
19.
Arterioscler Thromb Vasc Biol ; 32(10): 2444-51, 2012 Oct.
Article En | MEDLINE | ID: mdl-22904271

OBJECTIVE: Endothelium dysfunction is an initiating factor in atherosclerosis. A disintegrin and metalloproteinase 15 (ADAM 15) is a multidomain metalloprotease recently identified as a regulator of endothelial permeability. However, whether and how ADAM15 contributes to atherosclerosis remains unknown. METHODS AND RESULTS: Genetic ablation of ADAM15 in apolipoprotein E-deficient mice led to a significant reduction in aortic atherosclerotic lesion size (by 52%), plaque macrophage infiltration (by 69%), and smooth muscle cell deposition (by 82%). In vitro studies implicated endothelial-derived ADAM15 in barrier dysfunction and monocyte transmigration across mouse aortic and human umbilical vein endothelial cell monolayers. This role of ADAM15 depended on intact functioning of the cytoplasmic domain, as evidenced in experiments with site-directed mutagenesis targeting the metalloprotease active site (E349A), the disintegrin domain (Arginine-Glycine-Aspartic acid→Threonine-Aspartic acid-Aspartic acid), or the cytoplasmic tail. Further investigations revealed that ADAM15-induced barrier dysfunction was concomitant with dissociation of endothelial adherens junctions (vascular endothelial [VE]-cadherin/γ-catenin), an effect that was sensitive to Src family kinase inhibition. Through small interfering RNA-mediated knockdown of distinct Src family kinase members, c-Src and c-Yes were identified as important mediators of these junctional effects of ADAM15. CONCLUSIONS: These results suggest that endothelial cell-derived ADAM15, signaling through c-Src and c-Yes, contributes to atherosclerotic lesion development by disrupting adherens junction integrity and promoting monocyte transmigration.


ADAM Proteins/physiology , Atherosclerosis/physiopathology , Endothelium, Vascular/physiopathology , Membrane Proteins/physiology , Signal Transduction/physiology , src-Family Kinases/physiology , ADAM Proteins/drug effects , ADAM Proteins/genetics , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , CSK Tyrosine-Protein Kinase , Cell Movement/physiology , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/pathology , Humans , Membrane Proteins/drug effects , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/pathology , Monocytes/physiology , Proto-Oncogene Proteins c-yes/drug effects , Proto-Oncogene Proteins c-yes/genetics , Proto-Oncogene Proteins c-yes/physiology , RNA, Small Interfering/pharmacology , src-Family Kinases/drug effects , src-Family Kinases/genetics
20.
Vet Comp Oncol ; 10(3): 214-22, 2012 Sep.
Article En | MEDLINE | ID: mdl-22487216

Canine hemangiosarcoma (HSA) is an endothelial cell malignancy driven, in part, by activating mutations in receptor and non-receptor tyrosine kinases. Proteomics, Western blots and a tyrosine kinase inhibitor were used to elucidate activating mechanisms in HSA cell lines. Phosphotyrosine peptides from focal adhesion kinase (FAK) STAT3, Lyn, Fyn and other signal transduction kinases were identified by mass spectrometry. FAK was constitutively activated at tyrosine 397, the autophosphorylation site, and this was reversible with high concentrations of a FAK inhibitor. FAK inhibitor-14 suppressed migration and phosphorylation of FAK tyrosine 397 and tyrosines 576/577 and was cytotoxic to HSA cells suggesting FAK signalling may be an important contributor to canine HSA survival.


Dog Diseases/enzymology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Hemangiosarcoma/veterinary , Neoplasms, Vascular Tissue/veterinary , Phosphotyrosine/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Blotting, Western/veterinary , Cell Line, Tumor , Dog Diseases/drug therapy , Dogs , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Hemangiosarcoma/drug therapy , Hemangiosarcoma/enzymology , Mass Spectrometry/veterinary , Neoplasms, Vascular Tissue/drug therapy , Neoplasms, Vascular Tissue/enzymology , Proteomics/methods , Proto-Oncogene Proteins c-fyn/drug effects , Proto-Oncogene Proteins c-fyn/metabolism , STAT3 Transcription Factor/drug effects , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , src-Family Kinases/drug effects , src-Family Kinases/metabolism
...