Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Phytomedicine ; 128: 155319, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38518637

ABSTRACT

BACKGROUND: Myocardial infarction (MI) is one of the most deadly diseases in the world. Hyperoside (Hyp) has been shown to have a protective effect on cardiovascular function through various signaling pathways, but whether it can protect myocardial infarction by regulating JAK2/STAT3 signaling pathway is unknown. AIM OF THE STUDY: To investigate whether Hyp could protect the heart against myocardial infarction injury in mice by modulating JAK2/STAT3 signaling pathway and its potential mechanism. METHODS: In vivo experiments, the myocardial infarction model was established by ligating the left anterior descending coronary artery (LAD) of male C57BL/6 mice permanently. The mice were divided into seven groups: sham group, MI group, MI+Hyp (9 mg/kg), MI+Hyp (18 mg/kg) group, MI+Hyp (36 mg/kg) group, MI+Captopril group (15 mg/kg) group and MI+Hyp (36 mg/kg)+AG490 (7.5 mg/kg) group. Each group of animals were given different concentrations of hyperoside, positive control drug or inhibitor of JAK2/STAT3 singaling. After 14 days of administration, the electrocardiogram (ECG), echocardiography and serum myocardial injury markers were examined; Slices of mouse myocardial tissue were assessed for histopathological changes by HE, Masson and Sirius Red staining. TTC and TUNEL staining were used to evaluate the myocardial infarction area and cardiomyocytes apoptosis respectively. The expression of JAK2/STAT3 signaling pathway, apoptosis and autophagy-related proteins were detected by western blot. In vitro experiments, rat H9c2 cardiomyocytes were deprived of oxygen and glucose (OGD) to stimulate myocardial ischemia. The experiment was divided into seven groups: Control group, OGD group, OGD+Hyp (20 µM) group, OGD+Hyp (40 µM) group, OGD+Hyp (80 µM), OGD+Captopril (10 µM) group and OGD+Hyp (80 µM)+AG490 (100 µM) group. Myocardial cell damage and redox index were measured 12 h after OGD treatment. ROS content in cardiomyocytes was detected by immunofluorescence. Cardiomyocytes apoptosis was detected by flow cytometry. The expressions of JAK2/STAT3 signaling pathway-related proteins, apoptosis and autophagy related proteins were detected by western blot. RESULTS: In vivo, hyperoside could ameolirate ECG abnormality, increase cardiac function, reduce myocardial infarction size and significantly reduce myocardial fibrosis level and oxidation level. The experimental results in vitro showed that Hyp could reduce the ROS content in cardiomyocytes, decrease the level of oxidative stress and counteract the apoptosis induced by OGD injury . Both in vivo and in vitro experiments showed that hyperoside could increase phosphorylated JAK2 and STAT3, indicating that hyperoside could play a cardioprotective role by activating JAK2/STAT3 signaling pathway. It was also shown that hyperoside could increase the autophagy level of cardiomyocytes in vivo and in vitro. However the cardiomyocyte-protective effect of Hyp was abolished in combination with JAK2/ STAT3 signaling pathway inhibitor AG490. These results indicated that the protective effect of Hyp on cardiomyocyte injury was at least partially achieved through the activation of the JAK2/STAT3 signaling pathway. CONCLUSION: Hyp can significantly improve cardiac function, ameliorate myocardial hypertrophy and myocardial remodeling in MI mice. The mechanism may be related to improving mitochondrial autophagy of cardiomyocytes to maintain the advantage of autophagy, and blocking apoptosis pathway through phagocytosis, thus suppressing apoptosis level of cardiomyocytes. These effects of Hyp are achieved, at least in part, by activating the JAK2/STAT3 signaling pathway.


Subject(s)
Janus Kinase 2 , Mice, Inbred C57BL , Myocardial Infarction , Myocytes, Cardiac , Quercetin , Quercetin/analogs & derivatives , STAT3 Transcription Factor , Signal Transduction , Animals , STAT3 Transcription Factor/metabolism , Janus Kinase 2/metabolism , Myocardial Infarction/drug therapy , Male , Myocytes, Cardiac/drug effects , Signal Transduction/drug effects , Quercetin/pharmacology , Mice , Apoptosis/drug effects , Disease Models, Animal , Rats , Tyrphostins/pharmacology , Reactive Oxygen Species/metabolism
2.
Stem Cells Dev ; 28(21): 1424-1433, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31495275

ABSTRACT

Dome formation can occur in cultured tubular epithelial cells originating from various tissues, including the mammary gland and the kidney. The isolation and characterization of normal kidney epithelial stem cells that give rise to dome-forming tubular cells have never been reported. We attempted to isolate and characterize canine kidney epithelial stem cells using a simple cell culture method that we have previously used to isolate other adult human stem cells. Dome-forming kidney epithelial cells were derived from dissociated adult canine kidney tissues that were cultured in a modified keratinocyte serum-free medium supplemented with N-acetyl-l-cysteine, l-ascorbic acid 2-phosphate, nicotinamide, and fetal bovine serum. These cells exhibited high self-renewal capacity in long-term culture (growth for >13 months and 30 cumulative population doublings) and exhibited characteristics of stem cells, including (1) deficiency in gap junctional intercellular communication, (2) anchorage-independent growth, (3) expression of stem cell markers octamer-binding transcription factor 4 and SRY (sex determining region Y)-box 2, (4) expression of cell surface markers CD24 and CD133, and (5) multipotent differentiation into osteoblasts, adipocytes, chondrocytes, and dome-forming tubular cells. Most of these characteristics are shared by the well-known canine renal tubule-derived immortalized Madin-Darby Canine Kidney cell line. Furthermore, the putative canine kidney stem cells developed in this study formed budding tubule-like organoids on Matrigel and required high cell density (>4,000 cells/cm2) for sustained growth and confluency for dome formation. The signal transducer and activator of transcription-3 (STAT3) phosphorylation inhibitor, AG490, inhibited colony-forming efficiency and dome formation, whereas lipopolysaccharide, an activator of STAT3, increased colony-forming efficiency in a dose-dependent manner. These results are consistent with the hypothesis that high cell density induces STAT3 expression, which promotes both stem cell self-renewal and differentiation into tubular cells. Our novel cell culture method should be useful for the future development of normal human kidney stem cells for clinical applications and for studying mechanisms of nephrotoxicity.


Subject(s)
Epithelial Cells/cytology , Kidney Tubules/cytology , Multipotent Stem Cells/cytology , STAT3 Transcription Factor/metabolism , AC133 Antigen/metabolism , Animals , CD24 Antigen/metabolism , Cell Line , Cell- and Tissue-Based Therapy/methods , Dogs , Enzyme Inhibitors/pharmacology , Kidney Failure, Chronic/therapy , Lipopolysaccharides , Madin Darby Canine Kidney Cells , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Tyrphostins/pharmacology
3.
CNS Neurosci Ther ; 23(2): 106-118, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27671232

ABSTRACT

AIMS: Traumatic brain injury (TBI) is induced by complex primary and secondary mechanisms that give rise to cell death, inflammation, and neurological dysfunction. Understanding the mechanisms that drive neurological damage as well as those that promote repair can guide the development of therapeutic drugs for TBI. Kruppel-like factor 4 (KLF4) has been reported to negatively regulate axon regeneration of injured retinal ganglion cells (RGCs) through inhibition of JAK-STAT3 signaling. However, the role of KLF4 in TBI remains unreported. Reactive oxygen species (ROS)-induced neuronal death is a pathophysiological hallmark of TBI. METHODS: In this study, we used H2 O2 -treated RGCs in vitro and the optic nerve crush model in vivo to simulate neuronal damage in TBI. The function of KLF4 in RGC survival and axon regeneration in these models was investigated. In addition, the effects of KLF4 knockdown on neuronal damage after a brain impact that mimics moderate TBI were studied. RESULTS: The results show that H2 O2 induces p53-dependent apoptosis of RGCs in vitro through upregulation of KLF4. Additionally, KLF4 knockdown in vivo significantly enhances CNTF-induced axon regeneration of RGCs after optic nerve crush, and more importantly, prevents neuronal damage after a moderate brain impact in rats. Our Western blot analysis and immunoprecipitation assay results indicate that these effects of KLF4 knockdown are mediated by the p53 and JAK-STAT3 pathways. CONCLUSION: These findings provide evidence that KLF4 plays an important role in the pathophysiology of TBI. Blocking KLF4 may be a potential therapeutic strategy for the treatment of TBI, either alone or in combination with agents that target complementary mechanisms.


Subject(s)
Brain Injuries, Traumatic/pathology , Janus Kinases/metabolism , Kruppel-Like Transcription Factors/deficiency , Retinal Ganglion Cells/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cyclin D1/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Hydrogen Peroxide/pharmacology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Optic Nerve Injuries/pathology , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , STAT3 Transcription Factor/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Tyrphostins/pharmacology
4.
Mol Immunol ; 78: 65-78, 2016 10.
Article in English | MEDLINE | ID: mdl-27608299

ABSTRACT

Protein tyrosine kinases are key mediators of the signal transduction cascades that control expression of many genes involved in the induction of inflammation caused by arthritis. Here we investigate the effect of the tyrosine kinase inhibitor tyrphostin AG126 on a mouse model of adjuvant-induced arthritis (AIA). We report that when given at 5mg/kg i.p. every 48h from days 0-21, AG126 exerts potent anti-arthritic effects. Further, we investigated the role of AG126 on the key mediators of arthritic inflammation, namely, edema, arthritic score, presence of immunophenotypes including Foxp3+, CD4+Foxp3+, and CD25+Foxp3+ T regulatory (Treg) cells, as well as pro- and anti-inflammatory mediators. AG126 treatment significantly attenuated the severity of AIA and caused a substantial reduction in the percentage of CD2+, CD3+, CD4+, CD8+, CD23+, CD80+, CD86+ CD122+, CD195+, TCRß+, and GITR+ cells in whole blood. Moreover, administration of AG126 under arthritis-inducing conditions resulted in suppression of IL-17A+, IFN-γ+, CD4+ and CD25+ populations while causing an increase in the Foxp3+, CD4+Foxp3+, and CD25+Foxp3+ Treg populations in the spleen. In addition, RT-PCR analysis revealed increased expression of CD4, CD8, IL-17A, IFN-γ, TNF-α, and NF-κB p65 mRNAs and decreased IL-4 mRNA in the arthritic control (AC) mice, while treatment of animals with AG126 reversed these effects. Western blot analysis confirmed the decreased expression of IL-17, GITR, NF-κB p65 proteins and increased Foxp3 and IL-4 proteins following AG126 treatment of knee tissue. Thus, our findings provide new evidence that inhibition of protein tyrosine kinase activity decreases the progression of arthritis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Protein Kinase Inhibitors/pharmacology , T-Lymphocytes, Regulatory/drug effects , Animals , Blotting, Western , Cytokines/immunology , Female , Flow Cytometry , Immunohistochemistry , Mice , Mice, Inbred BALB C , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Regulatory/immunology , 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.
Biochem Biophys Res Commun ; 465(4): 719-24, 2015 Oct 02.
Article in English | MEDLINE | ID: mdl-26296463

ABSTRACT

We have recently discovered that melatonin, given acutely and directly to the isolated heart at the concentration found in wine, confers cardioprotection against ischemia-reperfusion (I/R). However, whether the presence of melatonin in wine contributes to the cardioprotective effect of chronic and moderate consumption of wine and its signalling mechanisms of protection are unknown. We therefore used both in vivo and in vitro models of I/R to investigate whether the presence of melatonin in red wine may contribute to the cardioprotective effect of chronic and moderate consumption of red wine. Wistar rats and C57black6 mice (WT) received drinking water supplemented daily with a moderate amount of red wine or melatonin given at the concentration found in the red wine. Rats were also pretreated with luzindole, a specific inhibitor of melatonin receptors 1 and 2 (2.3 mg/kg/day, intraperitoneally) or prazosin, a specific inhibitor of melatonin receptor type 3 (2.5 mg/kg/day, intraperitoneally). After 14 days, hearts were subjected to I/R in vivo or ex vivo. Red wine reduced the infarct size in both rats and WT mice (p < 0.001). Luzindole did not affect wine-induced cardioprotection, while prazosin reduced the infarct sparing effect of red wine (p < 0.05). Furthermore, red wine or melatonin failed to protect tumor necrosis factor alpha (TNF) receptor 2 knockout or cardiomyocyte specific signal transducer and activator of transcription 3 (STAT3) deficient mice (n.s. vs. control). Our novel findings suggest that the presence of melatonin in red wine contributes to the cardioprotective effect of chronic and moderate consumption of red wine against lethal I/R injuries. This effect is most likely mediated, at least in part, via melatonin receptor 3 and the activation of TNF and STAT3, both key players of the prosurvival and well described SAFE pathway.


Subject(s)
Cardiotonic Agents/administration & dosage , Melatonin/administration & dosage , Melatonin/metabolism , Receptors, Melatonin/metabolism , STAT3 Transcription Factor/metabolism , Wine/analysis , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/diet therapy , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Prazosin/pharmacology , Rats , Rats, Wistar , Receptor, Melatonin, MT1/antagonists & inhibitors , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/antagonists & inhibitors , Receptor, Melatonin, MT2/metabolism , Receptors, Melatonin/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type II/genetics , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/deficiency , STAT3 Transcription Factor/genetics , Tryptamines/pharmacology , Tyrphostins/pharmacology
7.
J Biomol Screen ; 20(1): 112-21, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25325257

ABSTRACT

Sirtuins are NAD(+)-dependent histone deacetylases (HDACs) that cleave off acetyl but also other acyl groups from the ϵ-amino group of lysines in histones and other substrate proteins. Five sirtuin isoforms are encoded in the genome of the parasitic pathogen Schistosoma mansoni. During its life cycle, S. mansoni undergoes drastic changes in phenotype that are associated with epigenetic modifications. Previous work showed strong effects of hSirt2 inhibitors on both worm life span and reproduction. Thus, we postulate smSirt2 as a new antiparasite target. We report both the optimization of a homogeneous fluorescence-based assay and the development of a new heterogeneous fluorescence-based assay to determine smSirt2 activity. The homogeneous assay uses a coumarin-labeled acetyl lysine derivative, and the heterogeneous version is using a biotinylated and fluorescence-labeled oligopeptide. Magnetic streptavidin-coated beads allow higher substrate loading per well than streptavidin-coated microtiter plates and make it possible to screen for inhibitors of either smSirt2 or its human isoform (hSirt2) for selectivity studies. We also present hits from a pilot screen with inhibitors showing an IC50 lower than 50 µM. Binding of the hits to their targets is rationalized by docking studies using a homology model of smSirt2.


Subject(s)
Fluorometry/methods , Schistosoma mansoni/enzymology , Sirtuin 2/metabolism , Animals , Binding Sites , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Epigenesis, Genetic/drug effects , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Conformation , Niacinamide/chemistry , Niacinamide/pharmacology , Protein Binding , Schistosoma mansoni/drug effects , Schistosoma mansoni/genetics , Sirtuin 2/antagonists & inhibitors , Sirtuin 2/genetics , Structure-Activity Relationship , Tyrphostins/chemistry , Tyrphostins/pharmacology
8.
Mol Brain ; 6: 45, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24152426

ABSTRACT

BACKGROUND: A luminex-based screen of cytokine expression in dorsal root ganglia (DRG) and nerve of type 1 diabetic rodents revealed interleukin-1 (IL-1α) and IL-1ß to be significantly depressed. We, therefore, tested the hypothesis that impaired IL-1α and IL-1ß expression in DRG may contribute to aberrant axon regeneration and plasticity seen in diabetic sensory neuropathy. In addition, we determined if these cytokines could optimize mitochondrial bioenergetics since mitochondrial dysfunction is a key etiological factor in diabetic neuropathy. RESULTS: Cytokines IL-1α and IL-1ß were reduced 2-fold (p<0.05) in DRG and/or nerve of 2 and 5 month streptozotocin (STZ)-diabetic rats. IL-2 and IL-10 were unchanged. IL-1α and IL-1ß induced similar 2 to 3-fold increases in neurite outgrowth in cultures derived from control or diabetic rats (p<0.05). STAT3 phosphorylation on Tyr705 or Ser727 was depressed in DRG from STZ-diabetic mice and treatment of cultures derived from STZ-diabetic rats with IL-1ß for 30 min raised phosphorylation of STAT3 on Tyr705 and Ser727 by 1.5 to 2-fold (p<0.05). shRNA-based or AG490 inhibition of STAT3 activity or shRNA blockade of endogenous IL-1ß expression completely blocked neurite outgrowth. Cultured neurons derived from STZ-diabetic mice were treated for 24 hr with IL-1ß and maximal oxygen consumption rate and spare respiratory capacity, both key measures of bioenergetic fidelity that were depressed in diabetic compared with control neurons, were enhanced 2-fold. This effect was blocked by AG490. CONCLUSIONS: Endogenous synthesis of IL-1ß is diminished in nerve tissue in type 1 diabetes and we propose this defect triggers reduced STAT3 signaling and mitochondrial function leading to sup-optimal axonal regeneration and plasticity.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Energy Metabolism , Interleukin-1beta/metabolism , Janus Kinases/metabolism , Mitochondria/metabolism , Neurites/metabolism , STAT3 Transcription Factor/metabolism , Aging/metabolism , Animals , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Energy Metabolism/drug effects , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Immunohistochemistry , Interleukin-1alpha/metabolism , Male , Mice , Mitochondria/drug effects , Mitochondria/enzymology , Neurites/drug effects , Neurites/enzymology , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Rats , Rats, Sprague-Dawley , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Signal Transduction/drug effects , Streptozocin , Tyrphostins/pharmacology
9.
Mol Med Rep ; 8(3): 891-6, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23877734

ABSTRACT

Sorghum is a principal cereal food in a number of parts of the world and is critical in folk medicine in Asia and Africa. However, its effects on bone are unknown. Growth hormone (GH) is a regulator of bone growth and bone metabolism. GH activates several signaling pathways, including the Janus kinase (Jak)/signal transducer and activator of transcription (STAT) pathways, thereby regulating expression of genes, including insulin­like growth factor (IGF)­1. Bone morphogenetic proteins (BMPs) induce the differentiation of cells of the osteoblastic lineage, increasing the pool of IGF­1 target cells, the mature osteoblasts. In the present study, the effects of Hwanggeumchal sorghum extracts (HSE) on GH signaling via the Jak/STAT pathway in osteoblasts were investigated. HSE was not observed to be toxic to osteoblastic cells and increased the expression of BMP7 and GH­related proteins, including STAT5B, p­STAT5B, IGF­1 receptor (IGF-1R), growth receptor hormone (GHR) and Jak2 in MC3T3­E1 cells. In addition, HSE increased BMP7 and GHR mRNA expression in MC3T3­E1 cells. The expression of HSE­induced BMP7 and GHR was inhibited by AG490, a Jak2 kinase inhibitor. The observations indicate that HSE­induced signaling is similar to GH signaling via the GHR­Jak2 signaling axis. Using small interference RNA (siRNA) analysis, STAT5B was found to play an essential role in HSE­induced BMP7 and GH signaling in MC3T3­E1 cells. Results of the current study indicate that HSE promotes bone growth through activation of STAT5B.


Subject(s)
Bone Morphogenetic Protein 7/metabolism , Growth Hormone/metabolism , Janus Kinase 2/metabolism , Plant Extracts/toxicity , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Sorghum/metabolism , Animals , Bone Morphogenetic Protein 7/genetics , Cell Differentiation , Cell Line , Cell Lineage , Gene Expression/drug effects , Janus Kinase 2/antagonists & inhibitors , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Plant Extracts/chemistry , RNA Interference , RNA, Small Interfering/metabolism , Receptor, IGF Type 1/metabolism , Receptors, Somatotropin/genetics , Receptors, Somatotropin/metabolism , STAT5 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/genetics , Sorghum/chemistry , Tyrphostins/pharmacology
10.
PLoS One ; 7(10): e47600, 2012.
Article in English | MEDLINE | ID: mdl-23094067

ABSTRACT

Choroidal neovascularisation (CNV) that occurs as a result of age-related macular degeneration (AMD) causes severe vision loss among elderly patients. The relationship between diabetes and CNV remains controversial. However, oxidative stress plays a critical role in the pathogenesis of both AMD and diabetes. In the present study, we investigated the influence of diabetes on experimentally induced CNV and on the underlying molecular mechanisms of CNV. CNV was induced via photocoagulation in the ocular fundi of mice with streptozotocin-induced diabetes. The effect of diabetes on the severity of CNV was measured. An immunofluorescence technique was used to determine the levels of oxidative DNA damage by anti-8-hydroxy-2-deoxyguanosine (8-OHdG) antibody, the protein expression of phosphorylated signal transducer and activator of transcription 3 (p-STAT3) and vascular endothelial growth factor (VEGF), in mice with CNV. The production of reactive oxygen species (ROS) in retinal pigment epithelial (RPE) cells that had been cultured under high glucose was quantitated using the 2',7'-dichlorofluorescein diacetate (DCFH-DA) method. p-STAT3 expression was examined using Western blot analysis. RT-PCR and ELISA processes were used to detect VEGF expression. Hyperglycaemia exacerbated the development of CNV in mice. Oxidative stress levels and the expression of p-STAT3 and VEGF were highly elevated both in mice and in cultured RPE cells. Treatment with the antioxidant compound N-acetyl-cysteine (NAC) rescued the severity of CNV in diabetic mice. NAC also inhibited the overexpression of p-STAT3 and VEGF in CNV and in RPE cells. The JAK-2/STAT3 pathway inhibitor AG490 blocked VEGF expression but had no effect on the production of ROS in vitro. These results suggest that hyperglycaemia promotes the development of CNV by inducing oxidative stress, which in turn activates STAT3 signalling in RPE cells. Antioxidant supplementation helped attenuate the development of CNV. Thus, our results reveal a potential strategy for the treatment and prevention of diseases involving CNV.


Subject(s)
Choroid/blood supply , Choroid/metabolism , Choroidal Neovascularization/metabolism , Hyperglycemia/pathology , Retinal Pigment Epithelium/metabolism , STAT3 Transcription Factor/genetics , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Choroid/drug effects , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/etiology , Choroidal Neovascularization/pathology , DNA Damage , Diabetes Mellitus, Experimental , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression/drug effects , Hyperglycemia/chemically induced , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Light Coagulation/adverse effects , Mice , Oxidative Stress/drug effects , Phosphorylation , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Retina/drug effects , Retina/metabolism , Retina/pathology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Severity of Illness Index , Signal Transduction/drug effects , Streptozocin , Tyrphostins/pharmacology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
11.
Int J Gynecol Cancer ; 22(9): 1457-62, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23099861

ABSTRACT

OBJECTIVE: To analyze the influence of epidermal growth factor receptor inhibitor AG1478 on the proliferation and epithelial-mesenchymal transition in endometrial carcinoma cells. MATERIALS AND METHODS: The inhibitory effect of different concentrations of AG1478 on the proliferation of endometrial carcinoma cells was detected by tetrazolium-based assay. Western blot was applied to investigate the influence of AG1478 on expressions of epithelium-cadherin, α-catenin, neural cadherin, vimentin, matrix metalloproteinase 9 (MMP9), and MMP2 protein in endometrial carcinoma cells. The influence of AG1478 on migration and invasion of endometrial carcinoma cells was examined by Transwell migration and invasion assay, respectively. RESULTS: AG1478 could suppress the proliferation of different endometrial carcinoma cells, and cells transfected with epidermal growth factor receptor were more sensitive (P < 0.05). The expression of an increase in epithelial marker proteins and a decrease in mesenchymal marker proteins, MMP9, MMP2 were observed in endometrial carcinoma cells after AG1478 treated. This effect was more obvious in cells transfected with epidermal growth factor receptor (P < 0.05). The migration and invasion ability of endometrial carcinoma cells were suppressed by AG1478, and Ishikawa cells transfected with epidermal growth factor receptor were demonstrated to be more sensitive to AG1478 (P < 0.05). CONCLUSIONS: Epidermal growth factor receptor inhibitor AG1478 could effectively inhibit the proliferation and epithelial-mesenchymal transition of endometrial carcinoma cells.


Subject(s)
Carcinoma/pathology , Endometrial Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Quinazolines/pharmacology , Tyrphostins/pharmacology , Carcinoma/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Endometrial Neoplasms/metabolism , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , Female , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness , Tumor Cells, Cultured
12.
Basic Res Cardiol ; 107(3): 263, 2012 May.
Article in English | MEDLINE | ID: mdl-22466958

ABSTRACT

In this study, we evaluated the effect of curcumin (Cur) post-treatment on isolated perfused rat hearts that had been subjected to a protocol of ischemia and reperfusion injury. We also examined whether the Janus kinase 2 and signal transducer and activator 3 of transcription (JAK2/STAT3) signaling pathway plays a role in the cardioprotective effects of Cur post-treatment. Isolated perfused rat hearts were subjected to 60 min of ischemia, followed by 60 min of reperfusion. The hearts were exposed to 1-µM Cur during the first 10 min of reperfusion in the absence or presence of the JAK kinase-specific inhibitor AG490 (AG, 1 µM). The Cur treatment conferred a cardioprotective effect, and the treated hearts demonstrated an improved post-ischemic cardiac functional recovery, a decreased myocardial infarct size and decreased lactate dehydrogenase release in the coronary flow, a reduced number of apoptotic cardiomyocytes, up-regulation of the anti-apoptotic protein Bcl2 and down-regulation of the pro-apoptotic protein Caspase3. AG blocked the Cur-mediated cardioprotection by inhibiting the JAK2/STAT3 signaling pathway, as reflected by the abrogation of the Cur-induced up-regulation of Bcl2 and down-regulation of Caspase3. The results suggest that Cur post-treatment can attenuate IR injury through the activation of the JAK2/STAT3 signaling pathway, which transmits a survival signal to the myocardium.


Subject(s)
Cardiotonic Agents/pharmacology , Curcumin/pharmacology , Janus Kinase 2/metabolism , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocardium/enzymology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Caspase 3/metabolism , In Vitro Techniques , Janus Kinase 2/antagonists & inhibitors , L-Lactate Dehydrogenase/metabolism , Male , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/pathology , Perfusion , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Recovery of Function , Time Factors , Tyrphostins/pharmacology , Ventricular Function, Left/drug effects , Ventricular Pressure/drug effects
13.
Eur J Med Chem ; 53: 133-40, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22520152

ABSTRACT

A new series of substituted quinazolin-4-(3H)-one-tyrphostin derivatives was prepared and screened for their cytotoxic activity against three tumor cell lines, namely human breast cancer cell line (MCF-7), human cervical cancer cell line (HeLa) and human hepatocellular liver carcinoma cell line (HepG2) using the colorimetric MTT assay. Among the current series, 10 compounds exhibited remarkable in vitro antiproliferative activity against the three tested cell lines with the IC(50) values ranging from 0.009 to 0.015 mM. All the compounds showed suitable drug like characteristics according to Lipinski's rule.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Quinazolines/chemistry , Tyrphostins/chemistry , Tyrphostins/pharmacology , Acrylamides/chemistry , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Drug Evaluation, Preclinical , Humans , Inhibitory Concentration 50 , Tyrphostins/chemical synthesis
14.
Reprod Fertil Dev ; 24(3): 482-9, 2012.
Article in English | MEDLINE | ID: mdl-22401280

ABSTRACT

Endometrial cysteine-rich protein 61 (CYR61, CCN1) is a growth factor-inducible gene whose expression is elevated during the proliferative phase of the menstrual cycle and which has been implicated in the pathogenesis of endometriosis. This study aimed to define the mediators of epidermal growth factor (EGF) signalling on CYR61 expression in spontaneously immortalised human endometrial epithelial cells (HES) as a model system. After 30 min of EGF treatment, the receptor was phosphorylated and internalised as well as mRNA CYR61 increased in HES cells. However, neither inhibition of C-terminal EGF receptor (EGFR)-phosphorylation nor blockage of the mitogen-activated proteinkinase/extracellular signal-regulated kinase (MAPK/ERK) pathway was able to reduce CYR61 levels. Surprisingly, the HES cells showed upregulation of CYR61 mRNA expression after inhibition of the MAPK/ERK pathway when treated with EGF. Specific inhibitor studies identified the contribution of Janus kinase 2 (JAK2) and the signal transducer and activator of transcription protein STAT3 to the regulation of CYR61 expression. The JAK2/STAT3 interaction contributed to the basal expression of CYR61 and mediated EGF-driven regulation of CYR61 after 30 and 120 min of treatment. In summary, EGF-mediated CYR61 upregulation in HES cells involves STAT3 and is counter-regulated by the EGFR/MAPK/ERK pathway.


Subject(s)
Cysteine-Rich Protein 61/genetics , Endometrium/drug effects , Epidermal Growth Factor/pharmacology , Janus Kinase 2/metabolism , STAT3 Transcription Factor/metabolism , Antineoplastic Agents/pharmacology , Cells, Cultured , Cysteine-Rich Protein 61/metabolism , Drug Evaluation, Preclinical , Endometrium/metabolism , Enzyme Activation/drug effects , Enzyme Activation/physiology , Epidermal Growth Factor/physiology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , ErbB Receptors/physiology , Female , Gene Expression Regulation/drug effects , Humans , Janus Kinase 2/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Quinazolines/pharmacology , STAT3 Transcription Factor/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Tyrphostins/pharmacology , Up-Regulation/drug effects
15.
Biochem Biophys Res Commun ; 420(1): 172-7, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22405821

ABSTRACT

Janus-activated kinase-2 JAK2 is activated by hyperosmotic shock and modifies the activity of several Na(+) coupled transporters. Carriers up-regulated by osmotic shock include the Na(+) coupled osmolyte transporter BGT1 (betaine/GABA transporter 1), which accomplishes the concentrative cellular uptake of γ-amino-butyric acid (GABA). The present study thus explored whether JAK2 participates in the regulation of BGT1 activity. To this end, cRNA encoding BGT1 was injected into Xenopus oocytes with or without cRNA encoding wild type JAK2, constitutively active (V617F)JAK2 or inactive (K882E)JAK2, and electrogenic GABA transport determined by dual electrode voltage clamp. In oocytes injected with cRNA encoding BGT1 but not in oocytes injected with water or with cRNA encoding JAK2 alone, the addition of 1mM GABA to the extracellular fluid generated an inward current (I(BGT)). In BGT1 expressing oocytes I(BGT) was significantly increased by coexpression of JAK2 or (V617F)JAK2, but not by coexpression of (K882E)JAK2. According to kinetic analysis coexpression of JAK2 increased the maximal I(BGT) without significantly modifying the concentration required for halfmaximal I(BGT) (K(M)). In oocytes expressing BGT1 and (V617F)JAK2 I(BGT) was gradually decreased by JAK2 inhibitor AG490 (40 µM). The decline of I(BGT) following disruption of carrier insertion with brefeldin A (5 µM) was similar in the absence and presence of the JAK2 inhibitor AG490 (40 µM). In conclusion, JAK2 is a novel regulator of the GABA transporter BGT1. The kinase up-regulates the carrier presumably by enhancing the insertion of carrier protein into the cell membrane.


Subject(s)
Carrier Proteins/biosynthesis , Janus Kinase 2/metabolism , Animals , Betaine/metabolism , Carrier Proteins/genetics , GABA Plasma Membrane Transport Proteins , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Mutation , Oocytes , Protein Kinase Inhibitors/pharmacology , RNA, Complementary/genetics , Tyrphostins/pharmacology , Up-Regulation , Xenopus , gamma-Aminobutyric Acid/metabolism
16.
Cancer Biol Ther ; 13(3): 130-7, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22231407

ABSTRACT

The inverse correlation between levels of dietary calcium and colorectal cancer (CRC) incidence has been extensively investigated. However, the impact of supplemental calcium on cancer therapy remains unknown. We used four models of CRC, Caco-2 and HCT116 human cancer cell lines and Apc (Min/+) and azoxymethane carcinogen-induced mouse models, to investigate the impact of a western-style diet low in calcium (0.05%) vs. a similar diet but supplemented with calcium (5%) on therapeutic targeting of the epidermal growth factor receptor (EGFR). We found that calcium supplementation combined with pharmacologic blockade of EGFR results in an additive effect on tumor growth inhibition in all models. Unexpectedly, the combined use of dietary calcium supplementation and EGFR inhibitors also resulted in elevated toxicity suggesting that careful consideration be given when combining dietary supplements with prescribed cancer therapies.


Subject(s)
Antineoplastic Agents/therapeutic use , Calcium/therapeutic use , Colonic Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Quinazolines/therapeutic use , Tyrphostins/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Azoxymethane , Calcium/pharmacology , Calcium/toxicity , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Dietary Supplements , Drug Screening Assays, Antitumor , Drug Synergism , Female , Humans , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , Quinazolines/pharmacology , Quinazolines/toxicity , Random Allocation , Tumor Burden/drug effects , Tyrphostins/pharmacology , Tyrphostins/toxicity
17.
Pigment Cell Melanoma Res ; 25(2): 131-43, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22252091

ABSTRACT

Small molecules complement genetic mutants and can be used to probe pigment cell biology by inhibiting specific proteins or pathways. Here, we present the results of a screen of active compounds for those that affect the processes of melanocyte and iridophore development in zebrafish and investigate the effects of a few of these compounds in further detail. We identified and confirmed 57 compounds that altered pigment cell patterning, number, survival, or differentiation. Additional tissue targets and toxicity of small molecules are also discussed. Given that the majority of cell types, including pigment cells, are conserved between zebrafish and other vertebrates, we present these chemicals as molecular tools to study developmental processes of pigment cells in living animals and emphasize the value of zebrafish as an in vivo system for testing the on- and off-target activities of clinically active drugs.


Subject(s)
Metabolic Networks and Pathways/drug effects , Pigmentation/drug effects , Small Molecule Libraries/analysis , Small Molecule Libraries/pharmacology , Zebrafish/metabolism , Animals , Cell Count , Chromatophores/cytology , Chromatophores/drug effects , Cyclooxygenase Inhibitors/pharmacology , Drug Evaluation, Preclinical , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Heterocyclic Compounds, 3-Ring/pharmacology , Melanocytes/cytology , Melanocytes/drug effects , Phenotype , Purines/pharmacology , Pyrimidinones/pharmacology , Roscovitine , Tyrphostins/pharmacology , Zebrafish/embryology
18.
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
19.
Cell Signal ; 23(12): 2076-85, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21855629

ABSTRACT

AG490 is a tyrosine kinase inhibitor with activity against Jak2 and apoptotic activity in specific leukemias. Due to its weak kinase inhibitory activity and poor pharmacology, we conducted a cell-based screen for derivatives with improved Jak2 inhibition and activity in animals. Two hits emerged from an initial small chemical library screen, and more detailed structure-activity relationship studies led to the development of WP1130 with 50-fold greater activity in suppressing Jak2-dependent cytokine signaling than AG490. However, WP1130 did not directly suppress Jak2 kinase activity, but mediated Jak2 ubiquitination resulting in its trafficking through HDAC6 to perinuclear aggresomes without cytokine stimulation or SOCS-1 induction. Jak2 primarily contained K63-linked ubiquitin polymers, and mutation of this lysine blocked Jak2 ubiquitination and mobilization in WP1130-treated cells. Further analysis demonstrated that WP1130, but not AG490, acts as a deubiquitinating enzyme (DUB) inhibitor, possibly through a Michael addition reaction. We conclude that chemical modification of AG490 resulted in development of a DUB inhibitor with activity against a DUB capable of modulating Jak2 ubiquitination, trafficking and signal transduction.


Subject(s)
Endopeptidases/metabolism , Janus Kinase 2/metabolism , Protease Inhibitors/pharmacology , Signal Transduction/drug effects , Tyrphostins/pharmacology , Ubiquitination , Amino Acid Substitution , Cell Line, Tumor , Cell Proliferation/drug effects , Cyanoacrylates , Drug Evaluation, Preclinical , Endopeptidases/chemistry , Enzyme Assays , HSP90 Heat-Shock Proteins/metabolism , Histone Deacetylase 6 , Histone Deacetylases/metabolism , Humans , Inhibitory Concentration 50 , Interleukin-6/pharmacology , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nitriles/pharmacology , Phosphorylation , Proteasome Inhibitors , Protein Transport/drug effects , Proteolysis , Pyridines/pharmacology , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism
20.
J Biomol Screen ; 16(8): 818-24, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21788393

ABSTRACT

The identification of compounds that specifically inhibit or kill cancer cells without affecting cells from healthy tissues is very challenging but very important for reducing the side effects of current cancer therapies. Hence, there is an urgent need for improved assays allowing the selectivity of a given compound to be monitored directly. The authors present an assay system based on the competitive co-cultivation of an excess of cancer cells with a small fraction of noncancer human indicator cells generating a fluorescence signal. In the absence of a specific anticancer compound, the cancer cells outgrow the indicator cells and abolish the fluorescence signal. In contrast, the presence of specific anticancer drugs (such as Tyrphostin-AG1478 or PLX4720) results in the selective growth of the indicator cells, giving rise to a strong fluorescence signal. Furthermore, the authors show that the nonspecific cytotoxic compound sodium azide kills both cancer and noncancer cells, and no fluorescence signal is obtained. Hence, this assay system favors the selection of compounds that specifically target cancer cells and decreases the probability of selecting nonspecific cytotoxic molecules. Z factors of up to 0.85 were obtained, indicating an excellent assay that can be used for high-throughput screening.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Coculture Techniques , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays , Kidney Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Fluorescence , Humans , Indoles/pharmacology , Kidney Neoplasms/pathology , Organ Specificity , Quinazolines , Reproducibility of Results , Sodium Azide/adverse effects , Sulfonamides/pharmacology , Tyrphostins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL