Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
Add more filters

Publication year range
1.
J Cell Biochem ; 120(6): 9147-9158, 2019 06.
Article in English | MEDLINE | ID: mdl-30582204

ABSTRACT

Interstitial cystitis (IC) is a heterogeneous syndrome with unknown etiology, and microRNAs (miRs) were found to be involved in IC. In our study, we aim to explore the role of miR-132 in the inflammatory response and detrusor fibrosis in IC through the Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathway in rat models. A rat model of IC was established and treated with the miR-132 mimic, miR-132 inhibitor, and/or JAK-STAT signaling pathway inhibitor AG490. Enzyme-linked immunosorbent assay was applied to measure the expression of interleukin (IL)-6, IL-10, interferon-γ (IFN-γ), and tumor necrosis factor-α (TNF-α), and intercellular adhesion molecule-1 (ICAM-1). The urodynamic test was performed to assess urodynamic parameters, and reverse transcription quantitative polymerase chain reaction and Western blot analysis for the expression of miR-132, STAT4, suppressors of cytokine signaling 3 (SOCS3), JAK2, vascular endothelial growth factor (VEGF), IFN-γ, and TNF-α. IC rats treated with miR-132 inhibitor and AG490 had decreased collagen fiber, inflammatory cell infiltration, and mast cells, lower expression of IL-6, IL-10, IFN-γ, TNF-α, ICAM-1, collagens I and III, and alleviated urodynamic parameters and decreased expression of STAT4, VEGF, JAK2, IFN-γ, TNF-α, and increased expression of SOCS3. Taken together, our data indicate that downregulation of miR-132 alleviates inflammatory response and detrusor fibrosis in IC via the inhibition of the JAK-STAT signaling pathway.


Subject(s)
Cystitis, Interstitial/metabolism , Inflammation/metabolism , Janus Kinases/metabolism , MicroRNAs/metabolism , Animals , Cystitis, Interstitial/drug therapy , Female , Humans , Immunohistochemistry , In Vitro Techniques , Inflammation/drug therapy , Janus Kinase 2/metabolism , MicroRNAs/genetics , Rats , Rats, Sprague-Dawley , STAT4 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/metabolism , Tyrphostins/therapeutic use , Vascular Endothelial Growth Factor A/metabolism
2.
Clin Exp Pharmacol Physiol ; 46(1): 75-85, 2019 01.
Article in English | MEDLINE | ID: mdl-30221384

ABSTRACT

Diabetic retinopathy (DR) is one of the most frequently occurring microvascular complications of diabetes. Recent evidence indicates that epidermal growth factor receptors (EGFRs) are critical pathogenic players in non-neoplastic diseases, including diabetic cardiomyopathy and DR. However, the precise pathogenic mechanism of EGFR in DR has yet to be fully understood. In this study, we developed a type 1 diabetic early-stage retinopathy mouse model using injections of streptozotocin and an oxygen-induced end-stage diabetic retinopathy (OIR) model characterized by hypoxia-induced revascularization. We tested the hypothesis that the pathogenesis of DR can be reduced by the classic EGFR inhibitor, AG1478, in the mouse models. Our data indicated that treatment of AG1478 prevented retinal dysfunction, and reduced impairment of retinal structures as well as mitochondrial structures in retinal blood vessels in diabetic mice. Furthermore, AG1478 reduced neovascular tufts formation but had no effects on revascularization at the avascular sites when compared to untreated littermates in the OIR model. Our findings provide strong evidence that EGFR critically promoted retinal dysfunction, retinal structural impairment, and retinal vascular abnormalities in models of DR. We conclude that EGFR can be a potential important therapeutic target for treatment of DR.


Subject(s)
Diabetic Retinopathy/pathology , Diabetic Retinopathy/physiopathology , ErbB Receptors/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Tyrphostins/pharmacology , Animals , Disease Progression , Female , Inflammation/pathology , Macrophages/cytology , Macrophages/drug effects , Male , Mice , Mitochondria/drug effects , Mitochondria/pathology , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Retinal Vessels/drug effects , Retinal Vessels/pathology , Tyrphostins/therapeutic use
3.
Pflugers Arch ; 470(3): 471-480, 2018 03.
Article in English | MEDLINE | ID: mdl-29288332

ABSTRACT

We previously reported that EGFR tyrosine kinase (EGFRtk) activity and endoplasmic reticulum (ER) stress are enhanced in type 2 diabetic (T2D) mice and cause vascular dysfunction. In the present study, we determined the in vivo contribution of EGFRtk and ER stress in acute myocardial infarction induced by acute ischemia (40 min)-reperfusion (24 h) (I/R) injury in T2D (db-/db-) mice. We treated db-/db- mice with EGFRtk inhibitor (AG1478, 10 mg/kg/day) for 2 weeks. Mice were then subjected to myocardial I/R injury. The db-/db- mice developed a significant infarct after I/R injury. The inhibition of EGFRtk significantly reduced the infarct size and ER stress induction. We also determined that the inhibition of ER stress (tauroursodeoxycholic acid, TUDCA, 150 mg/kg per day) in db-/db- significantly decrease the infarct size indicating that ER stress is a downstream mechanism to EGFRtk. Moreover, AG1478 and TUDCA reduced myocardium p38 and ERK1/2 MAP-kinases activity, and increased the activity of the pro-survival signaling cascade Akt. Additionally, the inhibition of EGFRtk and ER stress reduced cell apoptosis and the inflammation as indicated by the reduction in macrophages and neutrophil infiltration. We determined for the first time that the inhibition of EGFRtk protects T2D heart against I/R injury through ER stress-dependent mechanism. The cardioprotective effect of EGFRtk and ER stress inhibition involves the activation of survival pathway, and inhibition of apoptosis, and inflammation. Thus, targeting EGFRtk and ER stress has the potential for therapy to overcome myocardial infarction in T2D.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diabetic Cardiomyopathies/metabolism , Endoplasmic Reticulum Stress , ErbB Receptors/metabolism , Myocardial Infarction/metabolism , Animals , Apoptosis , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Diabetic Cardiomyopathies/drug therapy , ErbB Receptors/antagonists & inhibitors , MAP Kinase Signaling System , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Quinazolines/pharmacology , Quinazolines/therapeutic use , Taurochenodeoxycholic Acid/pharmacology , Taurochenodeoxycholic Acid/therapeutic use , Tyrphostins/pharmacology , Tyrphostins/therapeutic use
4.
Mol Carcinog ; 57(3): 429-439, 2018 03.
Article in English | MEDLINE | ID: mdl-29215754

ABSTRACT

Angiogenesis is an essential event in tumor growth and metastasis, and immune system also contributes to the tumor evasion. Emerging evidences have suggested the bidirectional link between angiogenesis and immunosuppression. Myeloid-derived suppressor cell (MDSC) is a kind of immunosuppressive cells and plays an important role in this process. However, the actual regulatory mechanisms of angiogenesis and MDSCs in head and neck squamous cell carcinoma (HNSCC) were unclear. In this study, through analyzing the immunohistochemistry staining of human HNSCC tissue microarray, we found that the microvascular density (MVD) was significantly increased in HNSCC patients. We also characterized angiogenic factors p-STAT3, VEGFA, CK2, and MDSCs marker CD11b in HNSCC tissue array, and found the close expression correlation among these markers. To determine the role of JAK2/STAT3 pathway in tumor microenvironment of HNSCC, we utilized AG490 (an inhibitor of JAK2/STAT3) for further research. Results showed that inhibition of JAK2/STAT3 suppressed angiogenesis by decreasing VEGFA and HIF1-α both in vitro and vivo. Moreover, in HNSCC transgenic mouse model, inhibiting JAK2/STAT3 not only suppressed angiogenesis but also reduced MDSCs in the tumor microenvironment through suppressing VEGFA and CK2. Our findings demonstrated the close relationship between angiogenesis and MDSCs in HNSCC, and inhibition of JAK2/STAT3 could reduce tumor-induced angiogenesis and decrease MDSCs.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Janus Kinase 2/antagonists & inhibitors , Myeloid-Derived Suppressor Cells/drug effects , Neovascularization, Pathologic/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Tyrphostins/therapeutic use , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Head and Neck Neoplasms/blood supply , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Human Umbilical Vein Endothelial Cells , Humans , Janus Kinase 2/metabolism , Mice, Transgenic , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Tyrphostins/pharmacology
5.
J Cell Mol Med ; 20(3): 482-94, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26762600

ABSTRACT

Cardiac hypertrophy is an important risk factor for heart failure. Epidermal growth factor receptor (EGFR) has been found to play a role in the pathogenesis of various cardiovascular diseases. The aim of this current study was to examine the role of EGFR in angiotensin II (Ang II)-induced cardiac hypertrophy and identify the underlying molecular mechanisms. In this study, we observed that both Ang II and EGF could increase the phospohorylation of EGFR and protein kinase B (AKT)/extracellular signal-regulated kinase (ERK), and then induce cell hypertrophy in H9c2 cells. Both pharmacological inhibitors and genetic silencing significantly reduced Ang II-induced EGFR signalling pathway activation, hypertrophic marker overexpression, and cell hypertrophy. In addition, our results showed that Ang II-induced EGFR activation is mediated by c-Src phosphorylation. In vivo, Ang II treatment significantly led to cardiac remodelling including cardiac hypertrophy, disorganization and fibrosis, accompanied by the activation of EGFR signalling pathway in the heart tissues, while all these molecular and pathological alterations were attenuated by the oral administration with EGFR inhibitors. In conclusion, the c-Src-dependent EGFR activation may play an important role in Ang II-induced cardiac hypertrophy, and inhibition of EGFR by specific molecules may be an effective strategy for the treatment of Ang II-associated cardiac diseases.


Subject(s)
Cardiomegaly/drug therapy , Cardiotonic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Quinazolines/pharmacology , Tyrphostins/pharmacology , Angiotensin II , Animals , Cardiomegaly/chemically induced , Cardiotonic Agents/therapeutic use , Cell Line , Drug Evaluation, Preclinical , Epidermal Growth Factor/physiology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Knockdown Techniques , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Quinazolines/therapeutic use , Rats , Tyrphostins/therapeutic use
6.
Genet Mol Res ; 14(1): 2450-60, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25867391

ABSTRACT

The aim of the present study was to investigate the anti-ovarian cancer effect of the inhibitor of signal transducer and activator of transcription 3 (STAT3), WP1066. Western blot was used to detect the phosphorylation of STAT3 in ovarian cancer cell line SKOV3 and cisplatin-resistant ovarian cancer cell line SKOV3/DDP. MTT and colony-forming assays were performed to evaluate the viability and growth of ovarian cancer cells. The apoptosis of ovarian cancer cells was determined by flow cytometry. The wound healing assay and Transwell assay were performed to examine the migration and invasion of ovarian cancer cells. WP1066 significantly inhibited the phosphorylation of STAT3 in SKOV3 and SKOV3/DDP cells. WP1066 treatment inhibited the proliferation and clonogenicity of both SKOV3 and SKOV3/DDP cells. After WP1066 treatment for 24 h, the apoptosis rates of SKOV3 and SKOV3/DDP cells were significantly increased compared with the control cells. After treatment with WP1066, the reduction of the wound gaps was significantly less in both SKOV3 and SKOV3/DDP cells. WP1066 also significantly inhibited the invasion capacity of SKOV3 and SKOV3/DDP cells compared with the control group. Treatment with WP1066 combined with cisplatin significantly increased proliferation inhibition and apoptosis in SKOV3 and SKOV3/ DDP cells compared with treatment with cisplatin alone. A synergistic action between WP1066 and cisplatin on the proliferation and apoptosis of ovarian cancer cells was determined. In conclusion, inhibition of STAT3 may suppress the proliferation, migration and invasion, induce apoptosis and enhance the chemosensitivity of ovarian cancer cells, indicating that STAT3 is a new therapeutic target of ovarian cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Ovarian Neoplasms/drug therapy , Pyridines/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Tyrphostins/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , Neoplasm Invasiveness , Phosphorylation , Pyridines/therapeutic use , Tyrphostins/therapeutic use
7.
Neurobiol Dis ; 62: 73-85, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24051278

ABSTRACT

Pilocarpine-induced status epilepticus (SE), which results in temporal lobe epilepsy (TLE) in rodents, activates the JAK/STAT pathway. In the current study, we evaluate whether brief exposure to a selective inhibitor of the JAK/STAT pathway (WP1066) early after the onset of SE affects the severity of SE or reduces later spontaneous seizure frequency via inhibition of STAT3-regulated gene transcription. Rats that received systemic WP1066 or vehicle at the onset of SE were continuously video-EEG monitored during SE and for one month to assess seizure frequency over time. Protein and/or mRNA levels for pSTAT3, and STAT3-regulated genes including: ICER, Gabra1, c-myc, mcl-1, cyclin D1, and bcl-xl were evaluated in WP1066 and vehicle-treated rats during stages of epileptogenesis to determine the acute effects of WP1066 administration on SE and chronic epilepsy. WP1066 (two 50mg/kg doses) administered within the first hour after onset of SE results in transient inhibition of pSTAT3 and long-term reduction in spontaneous seizure frequency. WP1066 alters the severity of chronic epilepsy without affecting SE or cell death. Early WP1066 administration reduces known downstream targets of STAT3 transcription 24h after SE including cyclin D1 and mcl-1 levels, known for their roles in cell-cycle progression and cell survival, respectively. These findings uncover a potential effect of the JAK/STAT pathway after brain injury that is physiologically important and may provide a new therapeutic target that can be harnessed for the prevention of epilepsy development and/or progression.


Subject(s)
Brain/physiopathology , Pyridines/therapeutic use , STAT3 Transcription Factor/antagonists & inhibitors , Status Epilepticus/drug therapy , Tyrphostins/therapeutic use , Animals , Brain/drug effects , Cell Death , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Disease Models, Animal , Electroencephalography , Hippocampus/drug effects , Hippocampus/metabolism , Phosphorylation , Pilocarpine , Pyridines/pharmacokinetics , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Seizures/drug therapy , Signal Transduction/drug effects , Status Epilepticus/chemically induced , Status Epilepticus/metabolism , Status Epilepticus/physiopathology , Tyrphostins/pharmacokinetics
8.
J Pharmacol Exp Ther ; 349(3): 458-69, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24696041

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) has been implicated as a hub for multiple oncogenic pathways. The constitutive activation of STAT3 is present in several cancers, including gliomas (GBMs), and is associated with poor therapeutic responses. Phosphorylation of STAT3 triggers its dimerization and nuclear transport, where it promotes the transcription of genes that stimulate tumor growth. In light of this role, inhibitors of the STAT3 pathway are attractive therapeutic targets for cancer. To this end, we evaluated the STAT3-inhibitory activities of three compounds (CPA-7 [trichloronitritodiammineplatinum(IV)], WP1066 [(S,E)-3-(6-bromopyridin-2-yl)-2-cyano-N-(1-phenylethyl)acrylamide, C17H14BrN3O], and ML116 [4-benzyl-1-{thieno[2,3-d]pyrimidin-4-yl}piperidine, C18H19N3S]) in cultured rodent and human glioma cells, including GBM cancer stem cells. Our results demonstrate a potent induction of growth arrest in GBM cells after drug treatment with a concomitant induction of cell death. Although these compounds were effective at inhibiting STAT3 phosphorylation, they also displayed variable dose-dependent inhibition of STAT1, STAT5, and nuclear factor κ light-chain enhancer of activated B cells. The therapeutic efficacy of these compounds was further evaluated in peripheral and intracranial mouse tumor models. Whereas CPA-7 elicited regression of peripheral tumors, both melanoma and GBM, its efficacy was not evident when the tumors were implanted within the brain. Our data suggest poor permeability of this compound to tumors located within the central nervous system. WP1066 and ML116 exhibited poor in vivo efficacy. In summary, CPA-7 constitutes a powerful anticancer agent in models of peripheral solid cancers. Our data strongly support further development of CPA-7-derived compounds with increased permeability to enhance their efficacy in primary and metastatic brain tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Glioma/drug therapy , Melanoma, Experimental/drug therapy , Melanoma, Experimental/secondary , STAT3 Transcription Factor/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chlorine Compounds/pharmacokinetics , Chlorine Compounds/pharmacology , Chlorine Compounds/therapeutic use , Drug Screening Assays, Antitumor , Glioma/metabolism , Glioma/pathology , HEK293 Cells , Heterocyclic Compounds, 2-Ring/pharmacokinetics , Heterocyclic Compounds, 2-Ring/pharmacology , Heterocyclic Compounds, 2-Ring/therapeutic use , Humans , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Piperidines/pharmacokinetics , Piperidines/pharmacology , Piperidines/therapeutic use , Platinum Compounds/pharmacokinetics , Platinum Compounds/pharmacology , Platinum Compounds/therapeutic use , Pyridines/pharmacokinetics , Pyridines/pharmacology , Pyridines/therapeutic use , STAT3 Transcription Factor/genetics , Small Molecule Libraries/pharmacokinetics , Small Molecule Libraries/therapeutic use , Tissue Distribution , Tyrphostins/pharmacokinetics , Tyrphostins/pharmacology , Tyrphostins/therapeutic use
9.
Exp Neurol ; 377: 114780, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38649091

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease characterized by dopaminergic neuron death and neuroinflammation. Emerging evidence points to the involvement of the transient receptor potential melastatin 2 (TRPM2) channel in neuron death and glial activation in several neurodegenerative diseases. However, the involvement of TRPM2 in PD and specifically its relation to the neuroinflammation aspect of the disease remains poorly understood. Here, we hypothesized that AG490, a TRPM2 inhibitor, can be used as a treatment in a mouse model of PD. Mice underwent stereotaxic surgery for 6-hydroxydopamine (6-OHDA) administration in the right striatum. Motor behavioral tests (apomorphine, cylinder, and rotarod) were performed on day 3 post-injection to confirm the PD model induction. AG490 was then daily injected i.p. between days 3 to 6 after surgery. On day 6, motor behavior was assessed again. Substantia nigra (SNc) and striatum (CPu) were collected for immunohistochemistry, immunoblotting, and RT-qPCR analysis on day 7. Our results revealed that AG490 post-treatment reduced motor behavior impairment and nigrostriatal neurodegeneration. In addition, the compound prevented TRPM2 upregulation and changes of the Akt/GSK-3ß/caspase-3 signaling pathway. The TRPM2 inhibition also avoids the glial morphology changes observed in the PD group. Remarkably, the morphometrical analysis revealed that the ameboid-shaped microglia, found in 6-OHDA-injected animals, were no longer present in the AG490-treated group. These results indicate that AG490 treatment can reduce dopaminergic neuronal death and suppress neuroinflammation in a PD mouse model. Inhibition of TRPM2 by AG490 could then represent a potential therapeutical strategy to be evaluated for PD treatment.


Subject(s)
Mice, Inbred C57BL , Neuroglia , TRPM Cation Channels , Tyrphostins , Animals , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/metabolism , Mice , Male , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Tyrphostins/pharmacology , Tyrphostins/therapeutic use , Disease Progression , Oxidopamine/toxicity , Disease Models, Animal , Nerve Degeneration/pathology , Nerve Degeneration/drug therapy , Parkinsonian Disorders/pathology , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/prevention & control , Substantia Nigra/drug effects , Substantia Nigra/pathology , Substantia Nigra/metabolism , Parkinson Disease/pathology , Parkinson Disease/metabolism , Parkinson Disease/drug therapy
10.
BMC Cancer ; 13: 297, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23777562

ABSTRACT

BACKGROUND: Over-expression of epidermal growth factor receptor (EGFR) or insulin-like growth factor-1 receptor (IGF-1R) have been shown to closely correlate with radioresistance of breast cancer cells. This study aimed to investigate the impact of co-inhibition of EGFR and IGF-1R on the radiosensitivity of two breast cancer cells with different profiles of EGFR and IGF-1R expression. METHODS: The MCF-7 (EGFR +/-, IGF-1R +++) and MDA-MB-468 (EGFR +++, IGF-1R +++) breast cancer cell lines were used. Radiosensitizing effects were determined by colony formation assay. Apoptosis and cell cycle distribution were measured by flow cytometry. Phospho-Akt and phospho-Erk1/2 were quantified by western blot. In vivo studies were conducted using MDA-MB-468 cells xenografted in nu/nu mice. RESULTS: In MDA-MB-468 cells, the inhibition of IGF-1R upregulated the p-EGFR expression. Either EGFR (AG1478) or IGF-1R inhibitor (AG1024) radiosensitized MDA-MB-468 cells. In MCF-7 cells, radiosensitivity was enhanced by AG1024, but not by AG1478. Synergistical radiosensitizing effect was observed by co-inhibition of EGFR and IGF-1R only in MDA-MB-468 cells with a DMF10% of 1.90. The co-inhibition plus irradiation significantly induced more apoptosis and arrested the cells at G0/G1 phase in MDA-MB-468 cells. Only co-inhibition of EGFR and IGF-1R synergistically diminished the expression of p-Akt and p-Erk1/2 in MDA-MB-468 cells. In vivo studies further verified the radiosensitizing effects by co-inhibition of both pathways in a MDA-MB-468 xenograft model. CONCLUSION: Our data suggested that co-inhibition of EGFR and IGF-1R synergistically radiosensitized breast cancer cells with both EGFR and IGF-1R high expression. The approach may have an important therapeutic implication in the treatment of breast cancer patients with high expression of EGFR and IGF-1R.


Subject(s)
Breast Neoplasms/radiotherapy , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , Quinazolines/pharmacology , Radiation Tolerance/drug effects , Receptor, IGF Type 1/antagonists & inhibitors , Tyrphostins/pharmacology , Analysis of Variance , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Drug Synergism , Enzyme Inhibitors/therapeutic use , ErbB Receptors/metabolism , Female , G1 Phase Cell Cycle Checkpoints/drug effects , G1 Phase Cell Cycle Checkpoints/radiation effects , Humans , MAP Kinase Signaling System/drug effects , MCF-7 Cells , Mice , Mice, Nude , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/therapeutic use , Tumor Stem Cell Assay , Tyrphostins/therapeutic use
11.
Nat Genet ; 28(1): 29-35, 2001 May.
Article in English | MEDLINE | ID: mdl-11326271

ABSTRACT

Hepatocellular carcinoma (HCC) is a major cause of cancer death, but the molecular mechanism for its development beyond its initiation has not been well characterized. Suppressor of cytokine signaling (SOCS-1; also known as JAB and SSI-1) switches cytokine signaling 'off' by means of its direct interaction with Janus kinase (JAK). We identified aberrant methylation in the CpG island of SOCS-1 that correlated with its transcription silencing in HCC cell lines. The incidence of aberrant methylation was 65% in the 26 human primary HCC tumor samples analyzed. Moreover, the restoration of SOCS-1 suppressed both growth rate and anchorage-independent growth of cells in which SOCS-1 was methylation-silenced and JAK2 was constitutively activated. This growth suppression was caused by apoptosis and was reproduced by AG490, a specific, chemical JAK2 inhibitor that reversed constitutive phosphorylation of STAT3 in SOCS-1 inactivated cells. The high prevalence of the aberrant SOCS-1 methylation and its growth suppression activity demonstrated the importance of the constitutive activation of the JAK/STAT pathway in the development of HCC. Our results also indicate therapeutic strategies for the treatment of HCC including use of SOCS-1 in gene therapy and inhibition of JAK2 by small molecules, such as AG490.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carrier Proteins/genetics , DNA Methylation , Gene Silencing , Genes, Tumor Suppressor , Intracellular Signaling Peptides and Proteins , Liver Neoplasms/genetics , Proto-Oncogene Proteins , Repressor Proteins , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/therapy , Carrier Proteins/metabolism , CpG Islands , DNA-Binding Proteins/metabolism , Genetic Therapy , Humans , Janus Kinase 2 , Liver Neoplasms/therapy , Molecular Sequence Data , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins , Trans-Activators/metabolism , Tyrphostins/therapeutic use
12.
Virology ; 588: 109900, 2023 11.
Article in English | MEDLINE | ID: mdl-37832343

ABSTRACT

Chikungunya virus (CHIKV) is a globally public health threat. There are currently no medications available to treat CHIKV infection. High-throughput screening of 419 kinase inhibitors was performed based on the cytopathic effect method, and six kinase inhibitors with reduced cytopathic effects, including tyrphostin AG879 (AG879), tyrphostin 9 (A9), sorafenib, sorafenib tosylate, regorafenib, and TAK-632, were identified. The anti-CHIKV activities of two receptor tyrosine kinase inhibitors, AG879 and A9, that have not been previously reported, were selected for further evaluation. The results indicated that 50% cytotoxic concentration (CC50) of AG879 and A9 in Vero cells were greater than 30 µM and 6.50 µM, respectively and 50% effective concentration (EC50) were 0.84 µM and 0.36 µM, respectively. The time-of-addition and time-of-removal assays illustrated that both AG879 and A9 function in the middle stage of CHIKV life cycle. Further, AG879 and A9 do not affect viral attachment; however, they inhibit viral RNA replication, and exhibit antiviral activity against CHIKV Eastern/Central/South African and Asian strains, Ross River virus and Sindbis virus in vitro.


Subject(s)
Antineoplastic Agents , Chikungunya Fever , Chikungunya virus , Animals , Chlorocebus aethiops , Humans , Chikungunya virus/genetics , Vero Cells , Tyrphostins/pharmacology , Tyrphostins/therapeutic use , Cell Line , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Virus Replication , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/pharmacology
13.
J Clin Immunol ; 32(5): 1038-47, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22661285

ABSTRACT

Previously, we have reported that the Jak-Stat signaling pathway is defective in NOD mice. In this study, prediabetic female NOD mice (4 weeks) were treated by intraperitoneal injection either with AG490 or DMSO three times per week for 4 consecutive weeks, followed by once a week for an additional 6 weeks. The onset of diabetes was attenuated in NOD mice treated with AG490 relative to DMSO treated control mice (p < 0.02). From an immunological standpoint, AG490 induced the expression of Foxp3 in CD4(+)CD25(-) T-cells and down-regulated expression of co-stimulatory molecules in dendritic cells (DC) both in vitro and in vivo. AG490 treated CD4+CD25- T-cells and DC in vitro, acquired regulatory functions; namely, the ability to suppress proliferation of a responding cell population in vitro. AG490 treatment resulted in significant reduction of blood glucose values and increased expression of PPARγ in splenocytes and markedly increased expression PPARγ2 but not PPARγ1 in adipocyte in vitro. Presence of multiple Stat5 DNA binding consensus sequences within the promoter region of the PPARγ gene in human and in mouse suggests that PPARγ is downstream to the Jak-Stat signaling pathway. This study highlights a critical role of the Jak-Stat signaling pathway in the pathogenesis of T1D and suggests that blocking the Jak-Stat signaling pathway by AG490 as a tyrosine kinase inhibitor may provide an effective means for preventing autoimmune T1D via both immunological and metabolic effects.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/prevention & control , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Tyrphostins/therapeutic use , Animals , Blood Glucose/analysis , Bone Marrow Cells/cytology , Cells, Cultured , Dendritic Cells , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/immunology , Female , Forkhead Transcription Factors/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , PPAR gamma/immunology , STAT Transcription Factors/immunology
14.
J Neuroinflammation ; 9: 178, 2012 Jul 23.
Article in English | MEDLINE | ID: mdl-22824323

ABSTRACT

BACKGROUND: Emerging evidence indicates that reactive microglia-initiated inflammatory responses are responsible for secondary damage after primary traumatic spinal cord injury (SCI); epidermal growth factor receptor (EGFR) signaling may be involved in cell activation. In this report, we investigate the influence of EGFR signaling inhibition on microglia activation, proinflammatory cytokine production, and the neuronal microenvironment after SCI. METHODS: Lipopolysaccharide-treated primary microglia/BV2 line cells and SCI rats were used as model systems. Both C225 and AG1478 were used to inhibit EGFR signaling activation. Cell activation and EGFR phosphorylation were observed after fluorescent staining and western blot. Production of interleukin-1 beta (IL-1 ß) and tumor necrosis factor alpha (TNF α) was tested by reverse transcription PCR and ELISA. Western blot was performed to semi-quantify the expression of EGFR/phospho-EGFR, and phosphorylation of Erk, JNK and p38 mitogen-activated protein kinases (MAPK). Wet-dry weight was compared to show tissue edema. Finally, axonal tracing and functional scoring were performed to show recovery of rats. RESULTS: EGFR phosphorylation was found to parallel microglia activation, while EGFR blockade inhibited activation-associated cell morphological changes and production of IL-1 ß and TNF α. EGFR blockade significantly downregulated the elevated MAPK activation after cell activation; selective MAPK inhibitors depressed production of cytokines to a certain degree, suggesting that MAPK mediates the depression of microglia activation brought about by EGFR inhibitors. Subsequently, seven-day continual infusion of C225 or AG1478 in rats: reduced the expression of phospho-EGFR, phosphorylation of Erk and p38 MAPK, and production of IL-1 ß and TNF α; lessened neuroinflammation-associated secondary damage, like microglia/astrocyte activation, tissue edema and glial scar/cavity formation; and enhanced axonal outgrowth and functional recovery. CONCLUSIONS: These findings indicate that inhibition of EGFR/MAPK suppresses microglia activation and associated cytokine production; reduces neuroinflammation-associated secondary damage, thus provides neuroprotection to SCI rats, suggesting that EGFR may be a therapeutic target, and C225 and AG1478 have potential for use in SCI treatment.


Subject(s)
ErbB Receptors/antagonists & inhibitors , ErbB Receptors/physiology , MAP Kinase Signaling System/physiology , Microglia/metabolism , Spinal Cord Injuries/metabolism , Animals , Animals, Newborn , Cells, Cultured , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , MAP Kinase Signaling System/drug effects , Male , Microglia/drug effects , Microglia/pathology , Quinazolines/pharmacology , Quinazolines/therapeutic use , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Tyrphostins/pharmacology , Tyrphostins/therapeutic use
15.
Phytother Res ; 26(1): 122-6, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21567511

ABSTRACT

Curcumin (diferuloylmethane) is a phenolic compound present in turmeric and is ingested daily in many parts of the world. Curcumin has been reported to cause inhibition on proliferation and induction of apoptosis in many human cancer cell lines, including non-small cell lung cancer cells (NSCLC). However, the clinical application of curcumin is restricted by its low bioavailability. In this report, it was observed that combined treatment of a low dosage of curcumin (5-10 µM) with a low concentration (0.1-2.5 µM) of small molecule inhibitors, including AG1478, AG1024, PD173074, LY294002 and caffeic acid phenethyl ester (CAPE) increased the growth inhibition in two human NSCLC cell lines: A549 and H1299 cells. The observation suggested that combined treatment of a low dosage of curcumin with inhibitors against epidermal growth factor receptor (EGFR), insulin-like growth factor 1 (IGF-1R), fibroblast growth factors receptor (FGFR), phosphatidylinositol 3-kinases (PI3K) or NF-κB signaling pathway may be a potential adjuvant therapy beneficial to NSCLC patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Curcuma/chemistry , Curcumin/therapeutic use , Enzyme Inhibitors/therapeutic use , Phytotherapy , Plant Extracts/therapeutic use , Antineoplastic Agents/pharmacology , Biological Availability , Caffeic Acids/pharmacology , Caffeic Acids/therapeutic use , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Chromones/pharmacology , Chromones/therapeutic use , Curcumin/pharmacology , Drug Therapy, Combination , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , Humans , Morpholines/pharmacology , Morpholines/therapeutic use , NF-kappa B/antagonists & inhibitors , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/pharmacology , Phenylethyl Alcohol/therapeutic use , Phosphoinositide-3 Kinase Inhibitors , Plant Extracts/pharmacology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Quinazolines/pharmacology , Quinazolines/therapeutic use , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Signal Transduction/drug effects , Somatomedins/antagonists & inhibitors , Tyrphostins/pharmacology , Tyrphostins/therapeutic use
16.
Int Immunopharmacol ; 113(Pt A): 109340, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36330910

ABSTRACT

Osteoarthritis (OA) is a complex chronic inflammatory disease characterized by articular degeneration and pain. Recent studies have identified interleukin 6 (IL-6) as a potential mediator leading to OA, but the therapeutic effects of inhibiting IL-6 signaling in intreating OA need to be further clarified. Here, we identified the intracellular signal transduction induced by recombinant IL-6 and focused on the impact of tyrphostin AG490 (a JAK2 inhibitor) on cartilage degeneration and OA pain. We found that IL-6 increased the inflammatory cytokines production and hypertrophic markers expression of primary mouse chondrocytes by activating JAK2/STAT3. Meanwhile, tyrphostin AG490 significantly attenuated articular degeneration and osteophyte formation in experimental mice with anterior cruciate ligament transection (ACLT) surgery. In vivo electrophysiological experiments showed that articular stimulation of IL-6 induced spinal hyperexcitability, which was prevented by coinjection of tyrphostin AG490. Specifically, compared with DMSO-treated ACLT mice, tyrphostin AG490 improved ambulate activity of mice and abolished the enhancement of serum bradykinin induced by IL-6. Together, we suggest that tyrphostin AG490 protected against progression of OA and improved OA prognosis by reducing cartilage degeneration and arthritis pain. Our findings provide further evidence for targeting IL-6 signaling in the treatment of OA.


Subject(s)
Cartilage, Articular , Osteoarthritis , Animals , Tyrphostins/pharmacology , Tyrphostins/therapeutic use , Interleukin-6/metabolism , Cartilage, Articular/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Chondrocytes , Pain/drug therapy , Pain/metabolism , Disease Models, Animal
17.
Sci Rep ; 12(1): 54, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34997096

ABSTRACT

Ulcerative colitis (UC) is a chronic inflammatory bowel disease. Several studies have demonstrated that α7 nicotinic acetylcholine receptors (α7nAChRs) exert anti-inflammatory effects on immune cells and nicotine suppress UC onset and relapse. Plasmacytoid dendritic cells (pDCs) reportedly accumulate in the colon of UC patients. Therefore, we investigated the pathophysiological roles of α7nAChRs on pDCs in the pathology of UC using oxazolone (OXZ)-induced Th2-type colitis with BALB/c mice. 2-deoxy-D-glucose, a central vagal stimulant suppressed OXZ colitis, and nicotine also ameliorated OXZ colitis with suppressing Th2 cytokines, which was reversed by α7nAChR antagonist methyllycaconitine. Additionally, α7nAChRs were expressed on pDCs, which were located very close to cholinergic nerve fibers in the colon of OXZ mice. Furthermore, nicotine suppressed CCL21-induced bone marrow-derived pDC migration due to Rac 1 inactivation, which was reversed by methyllycaconitine, a JAK2 inhibitor AG490 or caspase-3 inhibitor AZ-10417808. CCL21 was mainly expressed in the isolated lymphoid follicles (ILFs) of the colon during OXZ colitis. The therapeutic effect of cholinergic pathway on OXZ colitis probably through α7nAChRs on pDCs were attributed to the suppression of pDC migration toward the ILFs. Therefore, the activation of α7nAChRs has innovative therapeutic potential for the treatment of UC.


Subject(s)
Cholinergic Neurons/drug effects , Colitis, Ulcerative/drug therapy , Dendritic Cells/drug effects , Neuroimmunomodulation , Th2 Cells/metabolism , Aconitine/analogs & derivatives , Aconitine/pharmacology , Aconitine/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Caspase 3/metabolism , Caspase Inhibitors/pharmacology , Caspase Inhibitors/therapeutic use , Central Nervous System Stimulants/pharmacology , Central Nervous System Stimulants/therapeutic use , Colitis, Ulcerative/chemically induced , Colon/metabolism , Dendritic Cells/metabolism , Deoxyglucose/pharmacology , Deoxyglucose/therapeutic use , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Janus Kinase 2/metabolism , Mice, Inbred BALB C , Neuropeptides/metabolism , Nicotine/pharmacology , Nicotine/therapeutic use , Oxazolone/toxicity , STAT3 Transcription Factor/metabolism , Th2 Cells/drug effects , Tyrphostins/pharmacology , Tyrphostins/therapeutic use , Vagus Nerve/drug effects , alpha7 Nicotinic Acetylcholine Receptor/agonists , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors , alpha7 Nicotinic Acetylcholine Receptor/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism , rac1 GTP-Binding Protein/metabolism
18.
Antimicrob Agents Chemother ; 55(12): 5553-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21930873

ABSTRACT

We have previously reported that two receptor tyrosine kinase inhibitors (RTKIs), called AG879 and tyrphostin A9 (A9), can each block the replication of influenza A virus in cultured cells. In this study, we further characterized the in vitro antiviral efficacies and specificities of these agents. The 50% effective concentration (EC(50)) of each against influenza A was found to be in the high nanomolar range, and the selectivity index (SI = 50% cytotoxic concentration [CC(50)]/EC(50)) was determined to be >324 for AG879 and 50 for A9, indicating that therapeutically useful concentrations of each drug produce only low levels of cytotoxicity. Each compound showed efficacy against representative laboratory strains of both human influenza A (H1N1 or H3N2) and influenza B viruses. Importantly, no drug-resistant influenza virus strains emerged even after 25 viral passages in the presence of AG879, whereas viruses resistant to amantadine appeared after only 3 passages. AG879 and A9 each also exhibited potent inhibitory activity against a variety of other RNA and DNA viruses, including Sendai virus (Paramyxoviridae), herpes simplex virus (Herpesviridae), mouse hepatitis virus (Coronaviridae), and rhesus rotavirus (Reoviridae), but not against Pichinde virus (Arenaviridae). These results together suggest that RTKIs may be useful as therapeutics against viral pathogens, including but not limited to influenza, due to their high selectivity indices, low frequency of drug resistance, and broad-spectrum antiviral activities.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/drug effects , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Tyrphostins/pharmacology , Virus Replication/drug effects , Animals , Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Cell Line , Chick Embryo , Cricetinae , DNA Viruses/drug effects , DNA Viruses/physiology , Disease Models, Animal , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Humans , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/physiology , Influenza, Human/drug therapy , Influenza, Human/virology , Mice , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , RNA Viruses/drug effects , RNA Viruses/physiology , Rats , Treatment Outcome , Tyrphostins/chemistry , Tyrphostins/therapeutic use , Vero Cells
19.
Hepatology ; 52(3): 975-86, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20607690

ABSTRACT

UNLABELLED: Overexpression of epidermal growth factor receptor (ErbB1) and/or ErbB2 has been implicated in the pathogenesis of cholangiocarcinoma, suggesting that combined ErbB1/ErbB2 targeting might serve as a target-based therapeutic strategy for this highly lethal cancer. To test this strategy, we investigated targeting with the ErbB1 inhibitor tryphostin AG1517 and the ErbB2 inhibitor tryphostin AG879, in combination and alone, as well as with the dual ErbB1/ErbB2 inhibitor lapatinib, to assess the effectiveness of simultaneous targeting of ErbB1 and ErbB2 signaling over single inhibitor treatments in suppressing cholangiocarcinoma cell growth in vitro and the therapeutic efficacy of lapatinib in vivo. Our in vitro studies were carried out using rat (BDEneu and C611B) and human (HuCCT1 and TFK1) cholangiocarcinoma cell lines. The efficacy of lapatinib to significantly suppress liver tumor growth was tested in an orthotopic, syngeneic rat model of intrahepatic cholangiocarcinoma progression. Our results demonstrated that simultaneous targeting of ErbB1 and ErbB2 signaling was significantly more effective in suppressing the in vitro growth of both rat and human cholangiocarcinoma cells than individual receptor targeting. Lapatinib was an even more potent inhibitor of cholangiocarcinoma cell growth and inducer of apoptosis than either tryphostin when tested in vitro against these respective cholangiocarcinoma cell lines, regardless of differences in their levels of ErbB1 or ErbB2 protein expression and/or mechanism of activation. Lapatinib treatment also produced a significant suppression of intrahepatic cholangiocarcinoma growth when administered early to rats, but was without effect in inhibiting liver tumor growth in rats with more advanced tumors. CONCLUSION: Our findings suggest that simultaneous targeting of ErbB1 and ErbB2 could be a potentially selective strategy for cholangiocarcinoma therapy, but is likely to be ineffective by itself against advanced cancer.


Subject(s)
Bile Duct Neoplasms/drug therapy , Bile Ducts, Intrahepatic , Cholangiocarcinoma/drug therapy , ErbB Receptors/antagonists & inhibitors , Quinazolines/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Disease Models, Animal , ErbB Receptors/metabolism , Humans , Lapatinib , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Quinazolines/pharmacology , Rats , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , Tyrphostins/pharmacology , Tyrphostins/therapeutic use
20.
Cytokine ; 56(2): 343-50, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21843949

ABSTRACT

Previous studies indicate that administration of recombinant human erythropoietin (rhEPO) protects cortical neurons following traumatic brain injury (TBI). The mechanisms of rhEPO's neuroprotection are complex and interacting, including anti-apoptosis. Here we aim to demonstrate the role of janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) pathway on the anti-apoptotic effect of rhEPO in Feeney free falling TBI model. Activation of JAK2/STAT3 in pericontusional cortex was analyzed among rats in Sham, TBI, TBI+rhEPO, TBI+rhEPO+AG490 groups (rhEPO: 5000 U/kg day; JAK2 inhibitor AG490: 5 mg/kg day, intraperitoneal) through Western blotting, electrophoretic mobility shift assay. Bcl-2 and Bcl-xl expression (Q-PCR, Western blotting) and cell apoptosis (TUNEL) in pericontusional cortex were also detected in each group. As a result, we found that TBI could activate JAK2 and STAT3, and increase cell apoptosis in pericontusional cortex. RhEPO enhanced the expression of p-JAK2 and p-STAT3, up-regulated the mRNA and protein levels of Bcl-2 and Bcl-xl, followed by increased cell survival. Moreover, AG490 attenuated rhEPO's neuroprotection by down-regulating rhEPO-induced activation of JAK2/STAT3, and inhibiting Bcl-2 and Bcl-xl. These results suggest the essential role of JAK2/STAT3 pathway on the anti-apoptotic benefit of post-TBI rhEPO treatment.


Subject(s)
Apoptosis/drug effects , Brain Injuries/drug therapy , Erythropoietin/pharmacology , Animals , Base Sequence , Blotting, Western , Brain Injuries/metabolism , Brain Injuries/pathology , DNA Primers , Erythropoietin/therapeutic use , Humans , In Situ Nick-End Labeling , Male , Polymerase Chain Reaction , Rats , Rats, Wistar , Tyrphostins/pharmacology , Tyrphostins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL