Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Int J Mol Sci ; 23(9)2022 Apr 30.
Article in English | MEDLINE | ID: mdl-35563411

ABSTRACT

The emergence of the high correlation between type 2 diabetes and obesity with complicated conditions has led to the coinage of the term "diabesity". AMP-activated protein kinase (AMPK) activators and peroxisome proliferator-activated receptor (PPARγ) antagonists have shown therapeutic activity for diabesity, respectively. Hence, the discovery of compounds that activate AMPK as well as antagonize PPARγ may lead to the discovery of novel therapeutic agents for diabesity. In this study, the knockdown of PTPN6 activated AMPK and suppressed adipogenesis in 3T3-L1 cells. By screening a library of 1033 natural products against PTPN6, we found ethyl gallate to be the most selective inhibitor of PTPN6 (Ki = 3.4 µM). Subsequent assay identified ethyl gallate as the best PPARγ antagonist (IC50 = 5.4 µM) among the hit compounds inhibiting PTPN6. Ethyl gallate upregulated glucose uptake and downregulated adipogenesis in 3T3-L1 cells as anticipated. These results strongly suggest that ethyl gallate, which targets both PTPN6 and PPARγ, is a potent therapeutic candidate to combat diabesity.


Subject(s)
Diabetes Mellitus, Type 2 , Gallic Acid , PPAR gamma , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adipocytes/metabolism , Adipogenesis , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Gallic Acid/analogs & derivatives , Gallic Acid/pharmacology , Mice , Obesity/drug therapy , Obesity/metabolism , PPAR gamma/drug effects , PPAR gamma/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism
2.
Nat Metab ; 3(4): 513-522, 2021 04.
Article in English | MEDLINE | ID: mdl-33846641

ABSTRACT

Colchicine has served as a traditional medicine for millennia and remains widely used to treat inflammatory and other disorders. Colchicine binds tubulin and depolymerizes microtubules, but it remains unclear how this mechanism blocks myeloid cell recruitment to inflamed tissues. Here we show that colchicine inhibits myeloid cell activation via an indirect mechanism involving the release of hepatokines. We find that a safe dose of colchicine depolymerizes microtubules selectively in hepatocytes but not in circulating myeloid cells. Mechanistically, colchicine triggers Nrf2 activation in hepatocytes, leading to secretion of anti-inflammatory hepatokines, including growth differentiation factor 15 (GDF15). Nrf2 and GDF15 are required for the anti-inflammatory action of colchicine in vivo. Plasma from colchicine-treated mice inhibits inflammatory signalling in myeloid cells in a GDF15-dependent manner, by positive regulation of SHP-1 (PTPN6) phosphatase, although the precise molecular identities of colchicine-induced GDF15 and its receptor require further characterization. Our work shows that the efficacy and safety of colchicine depend on its selective action on hepatocytes, and reveals a new axis of liver-myeloid cell communication. Plasma GDF15 levels and myeloid cell SHP-1 activity may be useful pharmacodynamic biomarkers of colchicine action.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Colchicine/pharmacology , Cytokines/physiology , Liver/drug effects , Liver/metabolism , Myeloid Cells/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antioxidants/pharmacology , Colchicine/pharmacokinetics , Computer Simulation , Cytokines/biosynthesis , Growth Differentiation Factor 15/genetics , Hepatocytes/drug effects , Humans , Mice , Mice, Inbred C57BL , Microtubules/drug effects , Microtubules/metabolism , NF-E2-Related Factor 2/metabolism , Peritonitis/chemically induced , Peritonitis/prevention & control , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Signal Transduction/drug effects
3.
Asian Pac J Cancer Prev ; 22(3): 879-885, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33773553

ABSTRACT

OBJECTIVE: The natural compound, thymoquinone (TQ) has demonstrated potential anticancer properties in inhibiting cell proliferation and promoting apoptosis in myeloid leukemia cells, breast cancer cells, and others. However, the effect mechanism of TQ on AML cells still not fully understood. In this study, the authors examined the effects of TQ on the expression of JAK/STAT-negative regulator genes SOCS-1, SOCS-3, and SHP-1, and their consequences on cell proliferation and apoptosis in HL60 leukemia cells. METHODS: MTT and trypan blue exclusion tests were conducted to determine the 50% inhibitory concentration (IC50) and cell proliferation. FITC Annexin and Guava® reagent were used to study the cell apoptosis and examine the cell cycle phases, respectively. The expression of JAK/STAT-negative regulator genes, SOCS-1, SOCS-3, and SHP-1, was investigated using reverse transcriptase- quantitative PCR (RT-qPCR). RESULTS: TQ demonstrated a potential inhibition of HL60 cell proliferation and a significant increase in apoptotic cells in dose and time-dependent manner. TQ significantly induced cycle arrest at G0-G1 phase (P < 0.001) and enhanced the re-expression of JAK/STAT-negative regulator genes. CONCLUSION: TQ potentially inhibited HL60 cell proliferation and significantly increased apoptosis with re-expression of JAK/STAT-negative regulator genes suggesting that TQ could be a new therapeutic candidate for leukemia therapy.
.


Subject(s)
Apoptosis/drug effects , Benzoquinones/pharmacology , Cell Proliferation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Suppressor of Cytokine Signaling 1 Protein/drug effects , Suppressor of Cytokine Signaling 3 Protein/drug effects , HL-60 Cells , Humans , Inhibitory Concentration 50 , Janus Kinases , Leukemia, Promyelocytic, Acute/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , STAT Transcription Factors , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/genetics
4.
Aging (Albany NY) ; 13(1): 411-423, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33290262

ABSTRACT

As the first clinical proteasome inhibitor, Bortezomib (BTZ) has been reported to improve the outcome of lymphoma. However, due to the unstable property, low bioavailability, and hydrophobic properties of BTZ, it is needed to develop effective drug delivery systems to deliver BTZ into targeted cells or organs. Here we developed a bortezomib (BTZ)-loaded HMSNs (BTZ@HMSNs) system, which can sustain the release of BTZ in targeted tissues. In vitro assays showed that BTZ@HMSNs limited cell proliferation and augmented apoptosis of lymphoma SNK-1 cells. Moreover, BTZ@HMSNs significantly diminished migration and invasion of SNK-1 cells as compared with BTZ. In contrast to the upregulation of SHP-1, BTZ@HMSNs decreased the mRNA levels of c-Kit, NF-κB, and JAK1, which elicit oncogenic role in lymphoma development. Importantly, lymphoma mice model showed that BTZ@HMSNs significantly activated p53 signaling and reduced tumor volume and weight compared with free BTZ. Our data thus demonstrate that BTZ@HMSNs manifests improved tumor-suppressing effect in vitro and in vivo compared to free BTZ. We believe that HMSNs is a promising strategy for delivering therapeutic agents for cancer treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bortezomib/administration & dosage , Bortezomib/pharmacology , Cell Proliferation/drug effects , Lymphoma/drug therapy , Nanospheres , Animals , Cell Line, Tumor , Cell Movement/drug effects , Drug Carriers , Humans , In Vitro Techniques , Janus Kinase 1/drug effects , Janus Kinase 1/genetics , Lymphoma/genetics , Lymphoma/metabolism , Mice , Mice, Nude , NF-kappa B/drug effects , NF-kappa B/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-kit/genetics , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Silicon Dioxide , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
5.
Pharmacol Res ; 158: 104682, 2020 08.
Article in English | MEDLINE | ID: mdl-32035162

ABSTRACT

Advanced systemic mastocytosis is a rare and still untreatable disease. Blocking antibodies against inhibitory receptors, also known as "immune checkpoints", have revolutionized anti-cancer treatment. Inhibitory receptors are expressed not only on normal immune cells, including mast cells but also on neoplastic cells. Whether activation of inhibitory receptors through monoclonal antibodies can lead to tumor growth inhibition remains mostly unknown. Here we show that the inhibitory receptor Siglec-7 is expressed by primary neoplastic mast cells in patients with systemic mastocytosis and by mast cell leukemia cell lines. Activation of Siglec-7 by anti-Siglec-7 monoclonal antibody caused phosphorylation of Src homology region 2 domain-containing phosphatase-1 (SHP-1), reduced phosphorylation of KIT and induced growth inhibition in mast cell lines. In SCID-beige mice injected with either the human mast cell line HMC-1.1 and HMC-1.2 or with Siglec-7 transduced B cell lymphoma cells, anti-Siglec-7 monoclonal antibody reduced tumor growth by a mechanism involving Siglec-7 cytoplasmic domains in "preventive" and "treatment" settings. These data demonstrate that activation of Siglec-7 on mast cell lines can inhibit their growth in vitro and in vivo. This might pave the way to additional treatment strategies for mastocytosis.


Subject(s)
Lectins/agonists , Leukemia, Mast-Cell/drug therapy , Adult , Aged , Aged, 80 and over , Animals , Antibodies, Monoclonal/therapeutic use , Antigens, Differentiation, Myelomonocytic , Cell Line, Tumor , Cell Survival/drug effects , Female , Genes, src/drug effects , Humans , Leukemia, Mast-Cell/pathology , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/pathology , Male , Mastocytosis/drug therapy , Mice , Mice, SCID , Middle Aged , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Proto-Oncogene Proteins c-kit/drug effects , Xenograft Model Antitumor Assays
6.
Hum Mol Genet ; 28(13): 2120-2132, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30806670

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal, muscle degenerative disease causing premature death of affected children. DMD is characterized by mutations in the dystrophin gene that result in a loss of the dystrophin protein. Loss of dystrophin causes an associated reduction in proteins of the dystrophin glycoprotein complex, leading to contraction-induced sarcolemmal weakening, muscle tearing, fibrotic infiltration and rounds of degeneration and failed regeneration affecting satellite cell populations. The α7ß1 integrin has been implicated in increasing myogenic capacity of satellite cells, therefore restoring muscle viability, increasing muscle force and preserving muscle function in dystrophic mouse models. In this study, we show that a Food and Drug Administration (FDA)-approved small molecule, Sunitinib, is a potent α7 integrin enhancer capable of promoting myogenic regeneration by stimulating satellite cell activation and increasing myofiber fusion. Sunitinib exerts its regenerative effects via transient inhibition of SHP-2 and subsequent activation of the STAT3 pathway. Treatment of mdx mice with Sunitinib demonstrated decreased membrane leakiness and damage owing to myofiber regeneration and enhanced support at the extracellular matrix. The decreased myofiber damage translated into a significant increase in muscle force production. This study identifies an already FDA-approved compound, Sunitinib, as a possible DMD therapeutic with the potential to treat other muscular dystrophies in which there is defective muscle repair.


Subject(s)
Muscle, Skeletal/drug effects , Muscular Dystrophy, Duchenne/drug therapy , Myoblasts/drug effects , Sunitinib/therapeutic use , Animals , Cell Line , Disease Models, Animal , Disease Progression , Integrins/metabolism , Male , Mice , Mice, Inbred mdx , Muscle Development/drug effects , Muscle, Skeletal/metabolism , MyoD Protein/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Myogenin/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Regeneration , STAT3 Transcription Factor/drug effects , STAT3 Transcription Factor/metabolism , Satellite Cells, Skeletal Muscle/drug effects , Satellite Cells, Skeletal Muscle/metabolism , Sunitinib/pharmacology
7.
J Natl Cancer Inst ; 107(10)2015 Oct.
Article in English | MEDLINE | ID: mdl-26206949

ABSTRACT

BACKGROUND: Hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC) shows a higher incidence in men, mainly because of hepatitis B X (HBx)-mediated enhancement of androgen receptor (AR) activity. We aimed to examine this pathway in hepatocarcinogenesis and to identify drug(s) specifically blocking this carcinogenic event in the liver. METHODS: HBx transgenic mice that spontaneously develop HCC (n = 28-34 per group) were used, either by knockout of hepatic AR or by castration. Efficacy of several HCC-targeted drugs in suppressing HBx-induced AR activity was evaluated, and cellular factors mediating suppression were investigated in cultured cells. Tissue specificity of the candidate drug was validated using mouse tissues. Data were analyzed with Chi-square and Student's t tests. All statistical tests were two-sided. RESULTS: The androgen pathway was shown to be important in early stage hepatocarcinogenesis of HBx transgenic mice. The tumor incidence was decreased from 80% to 32% by AR knockout (P < .001) and from 90% to 25% by early castration (P < .001). Sorafenib markedly inhibited the HBx-enhanced AR activity through activating the SHP-1 phosphatase, which antagonized the activation of Akt/GSK3ß and c-Src by HBx. Moreover, SHP-1 protein level was much higher in the liver than in testis, which enabled sorafenib to inhibit aberrant AR activity in the HBx-expressing liver, while not affecting the physiological AR function in normal liver or testis. CONCLUSIONS: The androgen pathway may be a druggable target for the chemoprevention of HBV-related HCC, and sorafenib might be used as a tissue- and disease-specific regimen for the chemoprevention of HBV-related HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/prevention & control , Hepatitis B virus/metabolism , Hepatitis B/complications , Liver Neoplasms/prevention & control , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptors, Androgen/drug effects , Trans-Activators/metabolism , Age Factors , Animals , Carcinoma, Hepatocellular/virology , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Hepatitis B virus/pathogenicity , Incidence , Liver Neoplasms/virology , Male , Mice , Mice, Transgenic , Niacinamide/pharmacology , Orchiectomy , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Sorafenib , Trans-Activators/genetics , Viral Regulatory and Accessory Proteins
8.
Arthritis Rheumatol ; 67(7): 1766-77, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25833812

ABSTRACT

OBJECTIVE: Rheumatoid arthritis (RA), one of the most frequent chronic inflammatory rheumatic disorders, is characterized by the presence of autoantibodies and joint infiltration by activated immune cells, leading to cartilage and bone destruction. IgA occurs predominantly as monomers (mIgA) in plasma and regulates many cell responses through interaction with the Fcα receptor type I (FcαRI). FcαRI targeting by anti-FcαRI Fab inhibits activating receptors by inducing an inhibitory immunoreceptor tyrosine-based activation motif (ITAMi) configuration through SH2 domain-containing phosphatase 1 (SHP-1) recruitment. The aim of this study was to investigate the potential utility of mIgA for the treatment of arthritis by acting as an inducer of ITAMi signaling. METHODS: The effect of plasma-derived human mIgA on inhibition of multiple heterologous receptors was evaluated on FcαRI+ cell transfectants, blood phagocytes from healthy individuals, and synovial cells from RA patients. FcαRI-transgenic mice and wild-type mice treated with mIgA were studied in models of collagen antibody-induced arthritis (CAIA) and collagen-induced arthritis (CIA). The mice were assessed for development of arthritis using an arthritis score, and joint tissue samples were evaluated for the extent of leukocyte infiltration and expression of phosphatase. RESULTS: Treatment with mIgA impaired cell activation in an FcαRI-FcRγ-dependent manner, involving ITAMi signaling. Human mIgA or anti-FcαRI Fab were strongly effective in either preventing or attenuating CAIA or CIA in FcαRI-transgenic mice. Administration of mIgA markedly inhibited the recruitment of leukocytes to the inflamed joints of mice, which was associated with induction of SHP-1 phosphorylation in joint tissue cells. Moreover, mIgA reversed the state of inflammation in the synovial fluid of RA patients by inducing an ITAMi configuration. CONCLUSION: These results demonstrate a therapeutic potential of human mIgA in experimental arthritis. The findings support future clinical exploration of mIgA for the treatment of RA.


Subject(s)
Antigens, CD/physiology , Arthritis, Experimental/physiopathology , Immunoglobulin A/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/physiology , Receptors, Fc/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Animals , Antigens, CD/drug effects , Antigens, CD/genetics , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Case-Control Studies , Cell Line , Cells, Cultured , Chemotaxis/drug effects , Chemotaxis/physiology , Disease Models, Animal , Female , Humans , Immunoglobulin A/therapeutic use , In Vitro Techniques , Leukocytes/drug effects , Leukocytes/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Phagocytes/drug effects , Phagocytes/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Receptors, Fc/drug effects , Receptors, Fc/genetics , Synovial Membrane/drug effects , Synovial Membrane/pathology
9.
Hepatology ; 59(1): 190-201, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23908138

ABSTRACT

UNLABELLED: Sorafenib is the first approved targeted therapeutic reagent for hepatocellular carcinoma (HCC). Here, we report that Src homology region 2 (SH2) domain-containing phosphatase 1 (SHP-1) is a major target of sorafenib and generates a series of sorafenib derivatives to search for potent SHP-1 agonists that may act as better anti-HCC agents than sorafenib. Sorafenib increases SHP-1 activity by direct interaction and impairs the association between the N-SH2 domain and the catalytic protein tyrosine phosphatase domain of SHP-1. Deletion of the N-SH2 domain (dN1) or point mutation (D61A) of SHP-1 abolished the effect of sorafenib on SHP-1, phosphorylated signal transducer and activator of transcription 3 (p-STAT3), and apoptosis, suggesting that sorafenib may affect SHP-1 by triggering a conformational switch relieving its autoinhibition. Molecular docking of SHP-1/sorafenib complex confirmed our findings in HCC cells. Furthermore, novel sorafenib derivatives SC-43 and SC-40 displayed more potent anti-HCC activity than sorafenib, as measured by enhanced SHP-1 activity, inhibition of p-STAT3, and induction of apoptosis. SC-43 induced substantial apoptosis in sorafenib-resistant cells and showed better survival benefits than sorafenib in orthotopic HCC tumors. CONCLUSION: In this study, we identified SHP-1 as a major target of sorafenib. SC-43 and SC-40, potent SHP-1 agonists, showed better anti-HCC effects than sorafenib in vitro and in vivo. Further clinical investigation is warranted.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Catalytic Domain , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Humans , Mice , Models, Molecular , Niacinamide/pharmacology , Niacinamide/therapeutic use , Phenylurea Compounds/therapeutic use , Protein Tyrosine Phosphatase, Non-Receptor Type 6/chemistry , Random Allocation , STAT3 Transcription Factor/antagonists & inhibitors , Sorafenib , Xenograft Model Antitumor Assays
10.
Arch Pharm Res ; 34(7): 1201-8, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21811928

ABSTRACT

Ginsenosides, which are active compounds found in ginseng (Panax ginseng), are used as antidiabetic treatments. The aim of this study was to determine whether Rb2, a type of ginsenoside, regulates hepatic gluconeogenesis through AMP-activated protein kinase (AMPK) and the orphan nuclear receptor small heterodimer partner (SHP) in hyperlipidemic conditions used as an in vitro model of type 2 diabetes. Considering these results, we concluded that Rb2 may inhibit palmitate-induced gluconeogenesis via AMPK-induced SHP by relieving ER stress, a cause of gluconeogenesis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Ginsenosides/pharmacology , Hypoglycemic Agents/pharmacology , Phosphorylation/drug effects , Animals , Diabetes Mellitus, Type 2/drug therapy , Gene Silencing/drug effects , Ginsenosides/metabolism , Gluconeogenesis/drug effects , Glucose/analysis , Glucose/biosynthesis , Hepatocytes/drug effects , Hypoglycemic Agents/metabolism , Liver/drug effects , Liver/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , RNA/analysis , Rats , Transfection
11.
J Hepatol ; 55(5): 1041-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21354226

ABSTRACT

BACKGROUND & AIMS: Recently, we reported that sorafenib sensitizes hepatocellular carcinoma (HCC) cells to TRAIL through the inhibition of signal transducer and activator of transcription 3 (STAT3). Here, we report that sorafenib inhibits HCC via a kinase-independent mechanism: SHP-1 dependent STAT3 inactivation. METHODS: SC-1 is a sorafenib derivative that closely resembles sorafenib structurally but with no kinase inhibition activity. HCC cell lines (PLC5, Huh-7, Hep3B, and Sk-Hep1) were treated with sorafenib or SC-1 and apoptosis and signal transduction were analyzed. In vivo efficacy was determined in nude mice with Huh-7 xenografts. RESULTS: SC-1 showed similar effects to sorafenib on growth inhibition and apoptosis in all tested HCC cell lines. SC-1 down-regulated phosphorylation of phospho-STAT3 (p-STAT3) at tyrosine 705 in all tested HCC cells. Expression of STAT3-driven genes, including Cyclin D1 and Survivin, was also repressed by SC-1. Luciferase reporter assay confirmed the inhibition of transcriptional activity of STAT3 in both sorafenib-treated and SC-1-treated cells. Ectopic expression of STAT3 in PLC5 cells abolished apoptosis in SC-1-treated cells. Sorafenib and SC-1 up-regulated SHP-1 activity. Knockdown of SHP-1, but not SHP-2 or PTP-1B, by small interference RNA reduced apoptosis induced by SC-1. Finally, SC-1 reduced Huh-7 tumor growth significantly in vivo, which was associated with down-regulation of p-STAT3 and up-regulation of SHP-1 activity. CONCLUSIONS: STAT3 is a major kinase-independent target of sorafenib in HCC.


Subject(s)
Benzenesulfonates/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , STAT3 Transcription Factor/drug effects , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/enzymology , Cell Line, Tumor , Down-Regulation , Humans , Liver Neoplasms/enzymology , Mice , Mice, Nude , Niacinamide/analogs & derivatives , Phenylurea Compounds , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Proto-Oncogene Proteins c-raf/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Sorafenib
12.
Clin Immunol ; 133(1): 27-44, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19559654

ABSTRACT

Interferon-beta is a current treatment for multiple sclerosis (MS). Interferon-beta is thought to exert its therapeutic effects on MS by down-modulating the immune response by multiple potential pathways. Here, we document that treatment of MS patients with interferon beta-1a (Rebif) results in a significant increase in the levels and function of the protein tyrosine phosphatase SHP-1 in PBMCs. SHP-1 is a crucial negative regulator of cytokine signaling, inflammatory gene expression, and CNS demyelination as evidenced in mice deficient in SHP-1. In order to examine the functional significance of SHP-1 induction in MS PBMCs, we analyzed the activity of proinflammatory signaling molecules STAT1, STAT6, and NF-kappaB, which are known SHP-1 targets. Interferon-beta treatment in vivo resulted in decreased NF-kappaB and STAT6 activation and increased STAT1 activation. Further analysis in vitro showed that cultured PBMCs of MS patients and normal subjects had a significant SHP-1 induction following interferon-beta treatment that correlated with decreased NF-kappaB and STAT6 activation. Most importantly, experimental depletion of SHP-1 in cultured PBMCs abolished the anti-inflammatory effects of interferon-beta treatment, indicating that SHP-1 is a predominant mediator of interferon-beta activity. In conclusion, interferon-beta treatment upregulates SHP-1 expression resulting in decreased transcription factor activation and inflammatory gene expression important in MS pathogenesis.


Subject(s)
Interferon-beta/therapeutic use , Multiple Sclerosis/drug therapy , NF-kappa B/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , STAT1 Transcription Factor/metabolism , STAT6 Transcription Factor/metabolism , Adult , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cells, Cultured , Cytokines/blood , Female , Gene Silencing/immunology , Humans , Interferon beta-1a , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Male , Middle Aged , Multiple Sclerosis/immunology , NF-kappa B/antagonists & inhibitors , NF-kappa B/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , RNA, Small Interfering/immunology , RNA, Small Interfering/metabolism , STAT1 Transcription Factor/agonists , STAT1 Transcription Factor/immunology , STAT6 Transcription Factor/antagonists & inhibitors , STAT6 Transcription Factor/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
13.
Eur J Neurosci ; 29(2): 287-306, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19200235

ABSTRACT

Although the mechanisms underlying striatal neurodegeneration are poorly understood, we have shown that striatal pathogenesis may be initiated by high synaptic levels of extracellular dopamine (DA). Here we investigated in rat striatal primary neurons the mobilization of the mitogen-activated protein kinase (MAPK) signaling pathways after treatment with DA. Instead of observing an elevation of the archetypical pro-cytotoxic MAPKs, p-JNK and p-p38 MAPK, we found that DA, acting through D1 DA receptors, induced a sustained stimulation of the phosphorylated form of extracellular signal-regulated kinase (p-ERK) via a cAMP/protein kinase A (PKA)/Rap1/B-Raf / MAPK/ERK kinase (MEK) pathway. Blockade of D2 DA receptors, beta-adrenergic receptors or N-methyl-D-aspartate receptors with receptor-specific antagonists had no significant effect on this process. Activation of D1 DA receptors and PKA by DA caused phosphorylation and inactivation of the striatal-enriched tyrosine phosphatase, an important phosphatase for the dephosphorylation and subsequent inactivation of p-ERK in the striatum. Interestingly, p-ERK was primarily retained in the cytoplasm, with only low amounts translocated to the nucleus. The scaffold protein beta-arrestin2 interacted with both p-ERK and D1 DA receptor, triggering the cytosolic retention of p-ERK and inducing striatal neuronal apoptotic death. These data provide unique insight into a novel role of p-ERK in striatal neurodegeneration.


Subject(s)
Apoptosis/physiology , Corpus Striatum/metabolism , Dopamine/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/physiology , Nerve Degeneration/metabolism , Animals , Arrestins/metabolism , Basal Ganglia Diseases/metabolism , Basal Ganglia Diseases/physiopathology , Cells, Cultured , Corpus Striatum/physiopathology , Cyclic AMP-Dependent Protein Kinases/metabolism , Dopamine/pharmacology , Female , Nerve Degeneration/etiology , Nerve Degeneration/physiopathology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Phosphorylation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/drug effects , Receptors, Dopamine D1/metabolism , beta-Arrestins
14.
Mol Cancer Res ; 7(1): 118-28, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19147543

ABSTRACT

Activation of signal transducers and activators of transcription-3 (STAT-3) has been linked with survival, proliferation, chemoresistance, and angiogenesis of tumor cells, including human multiple myeloma (MM). Thus, agents that can suppress STAT3 activation have potential as cancer therapeutics. In our search for such agents, we identified acetyl-11-keto-beta-boswellic acid (AKBA), originally isolated from Boswellia serrata. Our results show that AKBA inhibited constitutive STAT3 activation in human MM cells. AKBA suppressed IL-6-induced STAT3 activation, and the inhibition was reversible. The phosphorylation of both Jak 2 and Src, constituents of the STAT3 pathway, was inhibited by AKBA. Interestingly, treatment of cells with pervanadate suppressed the effect of AKBA to inhibit the phosphorylation of STAT3, thus suggesting the involvement of a protein tyrosine phosphatase. We found that AKBA induced Src homology region 2 domain-containing phosphatase 1 (SHP-1), which may account for its role in dephosphorylation of STAT3. Moreover, deletion of the SHP-1 gene by small interfering RNA abolished the ability of AKBA to inhibit STAT3 activation. The inhibition of STAT3 activation by AKBA led to the suppression of gene products involved in proliferation (cyclin D1), survival (Bcl-2, Bcl-xL, and Mcl-1), and angiogenesis (VEGF). This effect correlated with the inhibition of proliferation and apoptosis in MM cells. Consistent with these results, overexpression of constitutive active STAT3 significantly reduced the AKBA-induced apoptosis. Overall, our results suggest that AKBA is a novel inhibitor of STAT3 activation and has potential in the treatment of cancer.


Subject(s)
Cell Survival/drug effects , Multiple Myeloma/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Transcription, Genetic/physiology , Triterpenes/pharmacology , Actins/genetics , Cell Line, Tumor , Flow Cytometry , Humans , Multiple Myeloma/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , RNA, Neoplasm/drug effects , RNA, Neoplasm/genetics , RNA, Small Interfering/genetics , STAT3 Transcription Factor/antagonists & inhibitors , Transcription, Genetic/drug effects
15.
Br J Pharmacol ; 153(5): 956-65, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18193074

ABSTRACT

BACKGROUND AND PURPOSE: Vascular endothelial growth factor (VEGF) is the most important proangiogenic protein. We have demonstrated that ATL-1, a synthetic analogue of aspirin-triggered lipoxin A(4), inhibits VEGF-induced endothelial cell (EC) migration. In the present study, we investigated the effects of ATL-1 in several other actions stimulated by VEGF. METHODS: Human umbilical vein ECs were treated with ATL-1 for 30 min before stimulation with VEGF. Cell proliferation was measured by thymidine incorporation. Adherent cells were determined by fluorescence intensity using a Multilabel counter. Expression and activity of matrix metalloproteinases (MMP) were analysed by western blot and zymography. KEY RESULTS: ATL-1 inhibited EC adhesion to fibronectin via interaction with its specific receptor. Furthermore, VEGF-induced MMP-9 activity and expression were reduced by pretreatment with ATL-1. Because the transcription factor NF-kappaB has been implicated in VEGF-mediated MMP expression and EC proliferation, we postulated that ATL-1 might modulate the NF-kappaB pathway and, indeed, ATL-1 inhibited NF-kappaB nuclear translocation. Pretreatment of EC with ATL-1 strongly decreased VEGF-dependent phosphorylation of phosphainositide 3-kinase (PI3-K) and extracellular signal-regulated kinase-2 (ERK-2), two signalling kinases involved in EC proliferation. Inhibition of VEGF-induced EC proliferation by ATL-1 was antagonized by sodium orthovanadate, suggesting that this inhibitory activity was mediated by a protein tyrosine phosphatase. This was confirmed by showing that ATL-1 inhibition of VEGF receptor-2 (VEGFR-2) phosphorylation correlates with SHP-1 association with VEGFR-2. CONCLUSIONS AND IMPLICATIONS: The synthetic 15-epi-lipoxin analogue, ATL-1, is a highly potent molecule exerting its effects on multiple steps of the VEGF-induced angiogenesis.


Subject(s)
Lipoxins/pharmacology , Neovascularization, Pathologic/prevention & control , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Vascular Endothelial Growth Factor A/metabolism , Active Transport, Cell Nucleus , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Humans , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , NF-kappa B/drug effects , NF-kappa B/metabolism , Neovascularization, Pathologic/physiopathology , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Umbilical Veins
16.
Atherosclerosis ; 191(1): 33-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-16725144

ABSTRACT

Vascular endothelial growth factor (VEGF) receptor-2 (KDR/flk-1) has a tyrosine kinase domain, and once activated, induces the autophosphorylation of the tyrosine residues, which is essential for angiogenesis. SHP-1, a cytoplasmic protein tyrosine phosphatase, plays a negative regulatory role in signal transduction pathways by dephosphorylation of the receptors to which it binds. Thus, therapeutic angiogenesis designed to inhibit expression of SHP-1 would be beneficial in hindlimb ischemia. In in vitro, the inhibition of SHP-1 by SHP-1 siRNA impaired the ability of TNF to block the tyrosine phosphorylation of KDR/flk-1 induced by VEGF and showed an increase in endothelial cell growth. In in vivo, SHP-1 mRNA, SHP-1 protein levels and VEGF were increased in a rat model of hindlimb ischemia. Upon injection to the ischemic adductor muscle, vector-based siRNA reduced SHP-1, increased phosphorylation of KDR/flk-1, and markedly increased capillary density. Our data demonstrated in vivo the potential use of siRNA targeting SHP-1 as therapy for peripheral ischemic diseases.


Subject(s)
Endothelial Cells/drug effects , Genetic Therapy , Ischemia/physiopathology , Neovascularization, Physiologic/drug effects , Peripheral Vascular Diseases/physiopathology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , RNA, Small Interfering/pharmacology , Animals , Cells, Cultured , Disease Models, Animal , Humans , Ischemia/therapy , Male , Peripheral Vascular Diseases/therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Rats , Rats, Wistar , Umbilical Veins , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...