Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Cancer Discov ; 12(7): 1742-1759, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35420681

ABSTRACT

Despite the popular use of dietary supplements during conventional cancer treatments, their impacts on the efficacies of prevalent immunotherapies, including immune-checkpoint therapy (ICT), are unknown. Surprisingly, our analyses of electronic health records revealed that ICT-treated patients with cancer who took vitamin E (VitE) had significantly improved survival. In mouse models, VitE increased ICT antitumor efficacy, which depended on dendritic cells (DC). VitE entered DCs via the SCARB1 receptor and restored tumor-associated DC functionality by directly binding to and inhibiting protein tyrosine phosphatase SHP1, a DC-intrinsic checkpoint. SHP1 inhibition, genetically or by VitE treatment, enhanced tumor antigen cross-presentation by DCs and DC-derived extracellular vesicles (DC-EV), triggering systemic antigen-specific T-cell antitumor immunity. Combining VitE with DC-recruiting cancer vaccines or immunogenic chemotherapies greatly boosted ICT efficacy in animals. Therefore, combining VitE supplement or SHP1-inhibited DCs/DC-EVs with DC-enrichment therapies could substantially augment T-cell antitumor immunity and enhance the efficacy of cancer immunotherapies. SIGNIFICANCE: The impacts of nutritional supplements on responses to immunotherapies remain unexplored. Our study revealed that dietary vitamin E binds to and inhibits DC checkpoint SHP1 to increase antigen presentation, prime antitumor T-cell immunity, and enhance immunotherapy efficacy. VitE-treated or SHP1-silenced DCs/DC-EVs could be developed as potent immunotherapies. This article is highlighted in the In This Issue feature, p. 1599.


Subject(s)
Cancer Vaccines , Neoplasms , Animals , Cancer Vaccines/therapeutic use , Dendritic Cells , Immunotherapy , Mice , Neoplasms/drug therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Vitamin E/metabolism
2.
Bioengineered ; 12(2): 10823-10836, 2021 12.
Article in English | MEDLINE | ID: mdl-34872449

ABSTRACT

To verify whether Ang-(1-7) produces an antagonistic effect on Ang II-mediated atrial remodeling. Ang II-induced HL-1 cell model and a rat model of Ang II-induced atrial remodeling were constructed and intervened with Ang II Ang-(1-7), AngII +Ang-(1-7), Ang II+ c-Src specific inhibitor (SU6656), and Ang II + Ang-(1-7) + SSG (SHP-1/2 specific inhibitor, stibogluconate), respectively. The systolic blood pressure of the rat caudal artery was detected. And trial fibrosis was detected by Picrosirius red staining and Masson's trichrome staining. Expressions of transforming growth factor-ß (TGF-ß), tissue inhibitor of metalloproteinases 1 (TIMP1), Matrix metalloproteinase 2 (MMP-2), connective tissue growth factor (CTGF), galectin-3, α-smooth muscle actin (α-SMA), and collagen I/III were subjected to qPCR and western blot. Furthermore, SHP-1 binding to c-Src was verified by co-immunoprecipitation (Co-IP). Results showed that the expressions of TGF-ß, TIMP1, MMP-2, CTGF, α-SMA, galectin-3, and collagen I were increased markedly in the Ang II intervention group, and the expressions of p-ERK1/2, p-Akt, and p-p38MAPK were also increased dramatically. Ang-(1-7) or SU6656 addition could inhibit the action of Ang II factor, thereby minimizing the expressions of the previously described genes and proteins. Simultaneously, SSG supplement reversed the antagonistic effect of Ang-(1-7) on Ang II, and the latter elevated the blood pressure and induced atrial fibrosis in rats. Ang-(1-7) could reverse the changes related to Ang II-induced atrial fibrosis in rats. In conclusion, Ang-(1-7) antagonized Ang II-induced atrial remodeling by regulating SHP-1 and c-Src, thereby affecting the MAPKs/Akt signaling pathway.


Subject(s)
Angiotensin II/metabolism , Angiotensin I/metabolism , Fibrosis/metabolism , Peptide Fragments/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , src Homology Domains/physiology , Animals , Matrix Metalloproteinase 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
3.
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
4.
Biomed Pharmacother ; 134: 111166, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33373915

ABSTRACT

Rheumatoid arthritis (RA) is a chronic autoimmune disease. Strong evidence supports that excessive activation of B cells plays a critical role in the pathogenesis of RA. Fc gamma receptor b (FcγRIIb) is the B cell inhibitory receptor and inhibits BCR (B cell receptor) signalling in part by selectively dephosphorylating CD19 which is considered a co-receptor for BCR and is essential for B cell activation. Our previous study demonstrated that a FcγRIIb I232T polymorphism presented a strong genetic link to RA and may lead to the excessive activation of B cells. Therefore, novel therapeutic strategies and drugs that can effectively inhibit the excessive activation of B cells by regulating the FcγRIIb are necessary for the treatment of RA. Therefore, we used Burkitt's lymphoma ST486 human B cells (lacking endogenous FcγRIIb) transfected with the 232Thr loss-of-function mutant to construct a FcγRIIb mutant cell line (ST486), and we demonstrated that YSTB treatment not only reduced proliferation and promoted apoptosis in ST486 cells but also did so in a dose-dependent manner. Furthermore, the intracellular Ca2+ flux of ST486 cells was decreased after treatment with YSTB, inhibiting the excessive activation of ST486 cells, and these effects correlated with the CD19/FcγRIIb-Lyn-SHP-1 pathways. Our data showed that YSTB treatment inhibited the expression of phosphorylated CD19 and upregulated the protein expression of FcγRIIb, Lyn, and SHP-1. Additionally, the CIA model was established to explore the anti-inflammatory and inhibitory effects of YSTB on bone destruction, and we found that YSTB decreased the paw oedema and arthritis index (AI) in CIA rats. It is worth mentioning that YSTB clearly decreased the AI earlier than methotrexate (MTX) (day 10 vs 16). Moreover, synovial hyperplasia, inflammatory cell infiltration and cartilage surface erosion in CIA rats were noticeably reduced after treatment with YSTB as evidenced by histopathological examination. Finally, we found that YSTB treatment suppressed bone erosion and joint space score (JNS) in CIA rats as evidenced by radiographic assessment. In summary, these data suggest that YSTB has great therapeutic potential for RA treatment.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/prevention & control , B-Lymphocytes/drug effects , Drugs, Chinese Herbal/pharmacology , Joints/drug effects , Lymphocyte Activation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptors, IgG/metabolism , src-Family Kinases/metabolism , Animals , Apoptosis/drug effects , Arthritis, Experimental/chemically induced , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Collagen Type II , Female , Humans , Joints/immunology , Joints/metabolism , Joints/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Rats, Wistar , Receptors, IgG/genetics , Signal Transduction , src-Family Kinases/genetics
5.
Arthritis Res Ther ; 22(1): 160, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32586377

ABSTRACT

BACKGROUND: The Src homology region 2 domain-containing phosphatase-1 (SHP-1) is known to exert negative regulatory effects on immune cell signaling. Mice with mutations in the Shp1 gene develop inflammatory skin disease and autoimmunity, but no arthritis. We sought to explore the role of SHP-1 in arthritis using an autoimmune mouse model of rheumatoid arthritis. We generated Shp1 transgenic (Shp1-Tg) mice to study the impact of SHP-1 overexpression on arthritis susceptibility and adaptive immune responses. METHODS: SHP-1 gene and protein expression as well as tyrosine phosphatase activity were evaluated in spleen cells of transgenic and wild type (WT) mice. WT and Shp1-Tg (homozygous or heterozygous for the transgene) mice were immunized with human cartilage proteoglycan (PG) in adjuvant, and arthritis symptoms were monitored. Protein tyrosine phosphorylation level, net cytokine secretion, and serum anti-human PG antibody titers were measured in immune cells from WT and Shp1-Tg mice. WT mice were treated with regorafenib orally to activate SHP-1 either before PG-induced arthritis (PGIA) symptoms developed (preventive treatment) or starting at an early stage of disease (therapeutic treatment). Data were statistically analyzed and graphs created using GraphPad Prism 8.0.2 software. RESULTS: SHP-1 expression and tyrosine phosphatase activity were elevated in both transgenic lines compared to WT mice. While all WT mice developed arthritis after immunization, none of the homozygous Shp1-Tg mice developed the disease. Heterozygous transgenic mice, which showed intermediate PGIA incidence, were selected for further investigation. We observed differences in interleukin-4 and interleukin-10 production in vitro, but serum anti-PG antibody levels were not different between the genotypes. We also found decreased tyrosine phosphorylation of several proteins of the JAK/STAT pathway in T cells from PG-immunized Shp1-Tg mice. Regorafenib administration to WT mice prevented the development of severe PGIA or reduced disease severity when started after disease onset. CONCLUSIONS: Resistance to arthritis in the presence of SHP-1 overexpression likely results from the impairment of tyrosine phosphorylation (deactivation) of key immune cell signaling proteins in the JAK/STAT pathway, due to the overwhelming tyrosine phosphatase activity of the enzyme in Shp1-Tg mice. Our study is the first to investigate the role of SHP-1 in autoimmune arthritis using animals overexpressing this phosphatase. Pharmacological activation of SHP-1 might be considered as a new approach to the treatment of autoimmune arthritis.


Subject(s)
Arthritis, Rheumatoid , Intracellular Signaling Peptides and Proteins , Animals , Arthritis, Rheumatoid/genetics , Mice , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatases/metabolism , Signal Transduction
6.
J Neuroimmunol ; 331: 46-57, 2019 06 15.
Article in English | MEDLINE | ID: mdl-29113698

ABSTRACT

Glycolysis and mitochondrial respiration are essential for oligodendrocyte metabolism in both the developing and adult CNS. Based on recent reports on the effects of the proinflammatory cytokine IFN-γ on metabolism and on oligodendrocytes, we addressed whether IFN-γ may affect oligodendrocyte bioenergetics in ways relevant to CNS disease. Oligodendrocytes of mice treated with IFN-γ showed significant reductions in aerobic glycolysis and mitochondrial respiration. As expected, IFN-γ treatment led to the induction of STAT1 in oligodendrocytes indicating active signaling into these cells. To determine the direct effects of IFN-γ on oligodendrocyte metabolism, cultured oligodendrocytes were treated with IFN-γ in vitro, which resulted in suppression of glycolysis similar to oligodendrocytes of animals treated with IFN-γ in vivo. Mice lacking SHP-1, a key regulator of IFN-γ and STAT1 signaling in CNS glia, had high constitutive levels of STAT1 and decreased aerobic glycolysis and mitochondrial respiration rates relative to wild type mouse oligodendrocytes. Together, these data show that IFN-γ and SHP-1 control oligodendrocyte bioenergetics in ways that may relate to the role of this cytokine in CNS disease.


Subject(s)
Energy Metabolism/drug effects , Interferon-gamma/pharmacology , Oligodendroglia/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/physiology , Animals , Cells, Cultured , Central Nervous System/pathology , Enzyme Activation/drug effects , Enzyme Induction/physiology , Female , Glycolysis/drug effects , Male , Mice , Mice, Inbred C3H , Mice, Neurologic Mutants , Oligodendroglia/metabolism , Oxidative Phosphorylation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/deficiency , STAT1 Transcription Factor/biosynthesis , STAT1 Transcription Factor/genetics , Signal Transduction/drug effects
7.
Part Fibre Toxicol ; 14(1): 53, 2017 Dec 12.
Article in English | MEDLINE | ID: mdl-29233151

ABSTRACT

BACKGROUND: Inhaled nanoparticles can deposit in the deep lung where they interact with pulmonary cells. Despite numerous studies on pulmonary nanotoxicity, detailed molecular mechanisms of specific nanomaterial-induced lung injury have yet to be identified. RESULTS: Using whole-body dynamic inhalation model, we studied the interactions between aluminum oxide nanoparticles (Al2O3 NPs) and the pulmonary system in vivo. We found that seven-day-exposure to Al2O3 NPs resulted in emphysema and small airway remodeling in murine lungs, accompanied by enhanced inflammation and apoptosis. Al2O3 NPs exposure led to suppression of PTPN6 and phosphorylation of STAT3, culminating in increased expression of the apoptotic marker PDCD4. Rescue of PTPN6 expression or application of a STAT3 inhibitor, effectively protected murine lungs from inflammation and apoptosis, as well as, in part, from the induction of chronic obstructive pulmonary disease (COPD)-like effects. CONCLUSION: In summary, our studies show that inhibition of PTPN6 plays a critical role in Al2O3 NPs-induced COPD-like lesions.


Subject(s)
Aluminum Oxide/toxicity , Lung/drug effects , Metal Nanoparticles/toxicity , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Pulmonary Disease, Chronic Obstructive/chemically induced , STAT3 Transcription Factor/metabolism , A549 Cells , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Disease Progression , Dose-Response Relationship, Drug , Humans , Inflammation Mediators/metabolism , Inhalation Exposure/adverse effects , Lung/enzymology , Lung/physiology , Male , Mice, Inbred C57BL , Phosphorylation , Pneumonia/chemically induced , Pneumonia/enzymology , Pneumonia/pathology , Pulmonary Disease, Chronic Obstructive/enzymology , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Disease, Chronic Obstructive/prevention & control , Pulmonary Emphysema/chemically induced , Pulmonary Emphysema/enzymology , Pulmonary Emphysema/pathology , RNA-Binding Proteins/metabolism , Signal Transduction/drug effects , Time Factors
8.
Int J Mol Med ; 40(5): 1566-1572, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28901387

ABSTRACT

Triptolide, an active component extracted from the medicinal plant Tripterygium wilfordii Hook F., has been used to treat various diseases, including lupus, cancer, rheumatoid arthritis and nephritic syndrome. The present study investigated the effects of triptolide on multiple myeloma using western blotting and an electrophoretic mobility shift assay. Triptolide was found to suppress the inducible and constitutive activation of signal transducer and activator of transcription 3 (STAT3), which is closely associated with inflammation and tumorigenesis. Triptolide also inhibited the DNA binding of STAT3. This correlated with the downregulation of Src kinase and Janus kinase 1 and 2, and with the upregulation of protein tyrosine phosphatase non­receptor type 6 (also known as SHP­1). In addition, triptolide downregulated the expression of the STAT3­regulated antiapoptotic (Bcl­xL and myeloid cell leukemia­1), proliferative (cyclin D1), and angiogenic (vascular endothelial growth factor) genes, suggesting that triptolide can induce apoptosis of tumor cells. These results suggest that triptolide may be a potential therapeutic anticancer agent for the prevention and treatment of multiple myeloma; thus further in­depth investigations into its efficacy and toxicity are warranted.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Diterpenes/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Phenanthrenes/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Antineoplastic Agents, Alkylating/chemistry , Cell Line, Tumor , Diterpenes/chemistry , Dose-Response Relationship, Drug , Enzyme Activation , Epoxy Compounds/chemistry , Epoxy Compounds/pharmacology , Humans , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Multiple Myeloma/pathology , Phenanthrenes/chemistry , Phosphorylation , Protein Binding , src-Family Kinases/metabolism
9.
Sci Rep ; 7(1): 1728, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28496142

ABSTRACT

This study aimed to investigate the role of src-homology protein tyrosine phosphatase-1 (SHP-1)-signal transducer and activator of transcription 3 (STAT3) pathway in liver fibrogenesis and the anti-fibrotic effect of SHP-1 agonist. The antifibrotic activity of SC-43, a sorafenib derivative with an enhanced SHP-1 activity, was evaluated in two fibrosis mouse models by carbon tetrachloride induction and bile duct ligation. Rat, human, and primary mouse hepatic stellate cells (HSCs) were used for mechanistic investigations. The results showed that SHP-1 protein primarily localized in fibrotic areas of human and mouse livers. SC-43 treatment reduced the activated HSCs and thus effectively prevented and regressed liver fibrosis in both fibrosis mouse models and improved mouse survival. In vitro studies revealed that SC-43 promoted HSC apoptosis, increased the SHP-1 activity and inhibited phospho-STAT3. The enhanced SHP-1 activity in HSCs significantly inhibited HSC proliferation, whereas SHP-1 inhibition rescued SC-43-induced HSC apoptosis. Furthermore, SC-43 interacted with the N-SH2 domain of SHP-1 to enhance the activity of SHP-1 as its antifibrotic mechanism. In conclusion, the SHP-1-STAT3 pathway is crucial in fibrogenesis. SC-43 significantly ameliorates liver fibrosis through SHP-1 upregulation. A SHP-1-targeted antifibrotic therapy may represent a druggable strategy for antifibrotic drug discovery.


Subject(s)
Liver Cirrhosis/drug therapy , Phenyl Ethers/therapeutic use , Phenylurea Compounds/therapeutic use , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Animals , Apoptosis/drug effects , Bile Ducts/pathology , Carbon Tetrachloride , Cell Line , Cell Proliferation/drug effects , Disease Models, Animal , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Humans , Ligation , Liver Cirrhosis/pathology , Liver Cirrhosis/prevention & control , Male , Mice, Inbred C57BL , Mutation/genetics , Phenyl Ethers/chemistry , Phenyl Ethers/pharmacology , Phenylurea Compounds/chemistry , Phenylurea Compounds/pharmacology , Protein Domains , Protein Tyrosine Phosphatase, Non-Receptor Type 6/chemistry , Rats , STAT3 Transcription Factor/metabolism , Sorafenib/chemistry , Sorafenib/pharmacology , Sorafenib/therapeutic use
10.
Molecules ; 22(2)2017 Feb 13.
Article in English | MEDLINE | ID: mdl-28208828

ABSTRACT

Ginkgolic acid C 17:1 (GAC 17:1) extracted from Ginkgo biloba leaves, has been previously reported to exhibit diverse antitumor effect(s) through modulation of several molecular targets in tumor cells, however the detailed mechanism(s) of its actions still remains to be elucidated. Signal transducer and activator of transcription 3 (STAT3) is an oncogenic transcription factor that regulates various critical functions involved in progression of diverse hematological malignancies, including multiple myeloma, therefore attenuating STAT3 activation may have a potential in cancer therapy. We determined the anti-tumor mechanism of GAC 17:1 with respect to its effect on STAT3 signaling pathway in multiple myeloma cell lines. We found that GAC 17:1 can inhibit constitutive activation of STAT3 through the abrogation of upstream JAK2, Src but not of JAK1 kinases in U266 cells and also found that GAC can suppress IL-6-induced STAT3 phosphorylation in MM.1S cells. Treatment of protein tyrosine phosphatase (PTP) inhibitor blocked suppression of STAT3 phosphorylation by GAC 17:1, thereby indicating a critical role for a PTP. We also demonstrate that GAC 17:1 can induce the substantial expression of PTEN and SHP-1 at both protein and mRNA level. Further, deletion of PTEN and SHP-1 genes by siRNA can repress the induction of PTEN and SHP-1, as well as abolished the inhibitory effect of drug on STAT3 phosphorylation. GAC 17:1 down-regulated the expression of STAT3 regulated gene products and induced apoptosis of tumor cells. Overall, GAC 17:1 was found to abrogate STAT3 signaling pathway and thus exert its anticancer effects against multiple myeloma cells.


Subject(s)
Ginkgo biloba/chemistry , PTEN Phosphohydrolase/metabolism , Plant Extracts/pharmacology , Plant Leaves/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , STAT3 Transcription Factor/agonists , Salicylates/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-6/metabolism , Membrane Potential, Mitochondrial/drug effects , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Phosphorylation/drug effects , Plant Extracts/chemistry , Protein Binding , Salicylates/chemistry
11.
Mol Oncol ; 11(3): 266-279, 2017 03.
Article in English | MEDLINE | ID: mdl-28084011

ABSTRACT

Recurrent triple-negative breast cancer (TNBC) needs new therapeutic targets. Src homology region 2 domain-containing phosphatase-1 (SHP-1) can act as a tumor suppressor by dephosphorylating oncogenic kinases. One major target of SHP-1 is STAT3, which is highly activated in TNBC. In this study, we tested a sorafenib analogue SC-60, which lacks angiokinase inhibition activity, but acts as a SHP-1 agonist, in TNBC cells. SC-60 inhibited proliferation and induced apoptosis by dephosphorylating STAT3 in both a dose- and time-dependent manner in TNBC cells (MDA-MB-231, MDA-MB-468, and HCC1937). By contrast, ectopic expression of STAT3 rescued the anticancer effect induced by SC-60. SC-60 also increased the SHP-1 activity, but this effect was inhibited when the N-SH2 domain (DN1) was deleted or with SHP-1 point mutation (D61A), implying that SHP-1 is the major target of SC-60 in TNBC. The use of SC-60 in combination with docetaxel synergized the anticancer effect induced by SC-60 through the SHP-1/STAT3 pathway in TNBC cells. Importantly, SC-60 also displayed a significant antitumor effect in an MDA-MB-468 xenograft model by modulating the SHP-1/STAT3 axis, indicating the anticancer potential of SC-60 in TNBC treatment. Targeting SHP-1/p-STAT3 and the potential combination of SHP-1 agonist with chemotherapeutic docetaxel is a feasible therapeutic strategy for TNBC.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Breast/drug effects , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , STAT3 Transcription Factor/metabolism , Taxoids/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast/metabolism , Breast/pathology , Cell Line, Tumor , Docetaxel , Drug Synergism , Female , Humans , Mice, Nude , Niacinamide/chemistry , Niacinamide/pharmacology , Niacinamide/therapeutic use , Phenylurea Compounds/chemistry , Phenylurea Compounds/pharmacology , Signal Transduction/drug effects , Sorafenib , Taxoids/pharmacology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
12.
Food Funct ; 7(8): 3468-79, 2016 Aug 10.
Article in English | MEDLINE | ID: mdl-27405813

ABSTRACT

Little is known about the chemical structure of purified extracellular polysaccharides from Phellinus sp., a fungal species with known medicinal properties. A combination of IR spectroscopy, methylation analysis and NMR were performed for the structural analysis of a purified extracellular polysaccharide derived from Phellinus sp. culture, denoted as SHP-1, along with an evaluation of the anti-aging effect in vivo of the polysaccharide supplementation. The structure of SHP-1 was established, with a backbone composed of →2,4)-α-d-glucopyranose-(1→ and →2)-ß-d-mannopyranose-(1→ and two terminal glucopyranose branches. Biochemical analysis from mammalian animal experiments demonstrated that SHP-1 possesses the ability to enhance antioxidant enzyme activities, such as catalase (CAT) and superoxide dismutase (SOD) activities, Trolox equivalent antioxidant capacity (TEAC) in serum of d-galactose-aged mice, while reducing lipofuscin levels, another indicator of cell aging, indicating a potential association with anti-aging activities in a dose dependent manner. This compound had a favourable influence on immune organ indices, and a marked amelioration ability of histopathological hepatic lesions such as necrosis, karyolysis and reduced inflammation and apoptosis in mouse hepatocytes. These results suggest that SHP-1 has strong antioxidant activities and a significant protective effect against oxidative stress or hepatotoxicity induced by d-galactose in mice and it could be developed as a food ingredient or a pharmaceutical to prevent many age-associated diseases such as major depressive disorder and hepatotoxicity. To our knowledge, this is the first report on the antioxidant effects of a novel purified exopolysaccharide derived from Phellinus sp.


Subject(s)
Aging/drug effects , Basidiomycota/chemistry , Polysaccharides/pharmacology , Animals , Apoptosis/drug effects , Catalase/metabolism , Depressive Disorder, Major/drug therapy , Disease Models, Animal , Female , Galactose , Hepatocytes/drug effects , Hepatocytes/metabolism , Inflammation/drug therapy , Magnetic Resonance Spectroscopy , Male , Mice , Oxidative Stress/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/pharmacology , Spectroscopy, Fourier Transform Infrared , Superoxide Dismutase/metabolism
13.
Phytother Res ; 30(4): 567-76, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27059688

ABSTRACT

Ginkgetin, a biflavone from Ginkgo biloba leaves, is known to exhibit antiinflammatory, antifungal, neuroprotective, and antitumor activities, but its precise mechanism of action has not been fully elucidated. Because the aberrant activation of STAT3 has been linked with regulation of inflammation, proliferation, invasion, and metastasis of tumors, we hypothesized that ginkgetin modulates the activation of STAT3 in tumor cells. We found that ginkgetin clearly suppressed constitutive phosphorylation of STAT3 through inhibition of the activation of upstream JAK1 and c-Src kinases and nuclear translocation of STAT3 on both A549 and FaDu cells. Treatment with sodium pervanadate reversed the ginkgetin-induced down-modulation of STAT3, thereby indicating a critical role for a PTP. We also found that ginkgetin strongly induced the expression of the SHP-1 and PTEN proteins and its mRNAs. Further, deletion of SHP-1 and PTEN genes by siRNA suppressed the induction of SHP-1 and PTEN, and reversed the inhibition of STAT3 activation. Ginkgetin induced apoptosis as characterized by an increased accumulation of cells in subG1 phase, positive Annexin V binding, loss of mitochondrial membrane potential, down-regulation of STAT3-regulated gene products, and cleavage of PARP. Overall, ginkgetin abrogates STAT3 signaling pathway through induction of SHP-1 and PTEN proteins, thus attenuating STAT3 phosphorylation and tumorigenesis.


Subject(s)
Apoptosis/drug effects , Biflavonoids/pharmacology , PTEN Phosphohydrolase/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , CSK Tyrosine-Protein Kinase , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , Humans , Janus Kinase 1/metabolism , Phosphorylation , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , src-Family Kinases/metabolism
14.
PLoS Biol ; 14(3): e1002401, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26942442

ABSTRACT

Demonstrations of both pro-apoptotic and pro-survival abilities of Fas (TNFRSF6/CD95/APO-1) have led to a shift from the exclusive "Fas apoptosis" to "Fas multisignals" paradigm and the acceptance that Fas-related therapies face a major challenge, as it remains unclear what determines the mode of Fas signaling. Through protein evolution analysis, which reveals unconventional substitutions of Fas tyrosine during divergent evolution, evolution-guided tyrosine-phosphorylated Fas proxy, and site-specific phosphorylation detection, we show that the Fas signaling outcome is determined by the tyrosine phosphorylation status of its death domain. The phosphorylation dominantly turns off the Fas-mediated apoptotic signal, while turning on the pro-survival signal. We show that while phosphorylations at Y232 and Y291 share some common functions, their contributions to Fas signaling differ at several levels. The findings that Fas tyrosine phosphorylation is regulated by Src family kinases (SFKs) and the phosphatase SHP-1 and that Y291 phosphorylation primes clathrin-dependent Fas endocytosis, which contributes to Fas pro-survival signaling, reveals for the first time the mechanistic link between SFK/SHP-1-dependent Fas tyrosine phosphorylation, internalization route, and signaling choice. We also demonstrate that levels of phosphorylated Y232 and Y291 differ among human cancer types and differentially respond to anticancer therapy, suggesting context-dependent involvement of Fas phosphorylation in cancer. This report provides a new insight into the control of TNF receptor multisignaling by receptor phosphorylation and its implication in cancer biology, which brings us a step closer to overcoming the challenge in handling Fas signaling in treatments of cancer as well as other pathologies such as autoimmune and degenerative diseases.


Subject(s)
Evolution, Molecular , Neoplasms/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , fas Receptor/metabolism , src-Family Kinases/metabolism , Amino Acid Sequence , Apoptosis , Endocytosis , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Protein Structure, Tertiary
15.
Cancer Lett ; 371(2): 205-13, 2016 Feb 28.
Article in English | MEDLINE | ID: mdl-26679051

ABSTRACT

Sorafenib is the first and currently the only standard treatment for advanced hepatocellular carcinoma (HCC). We previously developed a sorafenib derivative SC-43, which exhibits much more enhanced anti-HCC activity than sorafenib and also promotes apoptosis in sorafenib-resistant HCC cells. Herein, a novel "sorafenib plus" combination therapy was developed by coupling sorafenib treatment with SC-43. Both sorafenib and SC-43 are proven Src homology region 2 domain containing phosphatase 1 (SHP-1) agonists. The combined actions of sorafenib and SC-43 enhanced SHP-1 activity, which was associated with diminished STAT3-related signals and stronger expression of apoptotic genes above that of either drug alone, culminating in increased cell death. Decreased p-STAT3 signaling and tumor size, as well as increased SHP-1 activity were observed in mice receiving the combination therapy in a subcutaneous HCC model. More reduced orthotopic HCC tumor size and prolonged survival were also observed in mice in the combination treatment arm compared to mice in either of the monotherapy arms. These results in the preclinical setting pave the way for further clinical studies to treat unresectable HCC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular/drug therapy , Enzyme Activators/pharmacology , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenyl Ethers/pharmacology , Phenylurea Compounds/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Activation , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Mice, Nude , Niacinamide/pharmacology , Phosphorylation , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Sorafenib , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
16.
Pancreas ; 45(3): 409-19, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26646273

ABSTRACT

OBJECTIVES: Signal transducer and activator of transcription 3 (STAT3) is an oncogenic transcription factor implicated in carcinogenesis. Here, we investigated the role of morusin, the major prenylflavonoid, isolated from Chinese herbal medicine in abrogating the constitutive STAT3 activation in human pancreatic tumor cells. METHODS: The effect of morusin on STAT3 activation, associated protein kinases, STAT3-regulated gene products, cellular proliferation, and apoptosis was examined. RESULTS: Morusin specifically inhibited constitutive STAT3 activation both at tyrosine residue 705 and serine residue 727 in 4 pancreatic tumor cells. The inhibition of STAT3 was mediated through the suppression of activation of upstream JAK1, JAK2, and c-Src kinases. Morusin led to the accumulation of the cells in different phases of the cell cycle and caused induction of apoptosis and loss of mitochondrial membrane potential. Morusin downregulated the expression of various STAT3-regulated gene products; this correlated with induction of caspase-3 activation and anti-invasive effects. Treatment with the protein tyrosine phosphatase inhibitor pervanadate reversed the morusin-induced downregulation of STAT3, thereby suggesting the involvement of a protein tyrosine phosphatase. CONCLUSIONS: Morusin is a novel blocker of STAT3 activation and thus may have potential in negative regulation of growth and metastasis of pancreatic tumor cells.


Subject(s)
Apoptosis/drug effects , Cell Movement/drug effects , Flavonoids/pharmacology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Blotting, Western , CSK Tyrosine-Protein Kinase , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Reverse Transcriptase Polymerase Chain Reaction , src-Family Kinases/metabolism
17.
Pak J Pharm Sci ; 28(2): 421-4, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25730798

ABSTRACT

Protein tyrosine phosphatases (PTPs) are enzymes that catalyze protein tyrosine dephosphorylation of which Src homology phosphatase-1 (SHP-1) is one of the best-validated, a widely distributed intracellular tyrosine phosphatase that contains two SH2 domains. Down regulation of SHP-1 tyrosine phosphatases was significantly increased sensitivity to insulin in insulin signaling pathway. Through in vitro enzymatic reaction kinetics experiment, we found that the extract of Perilla stem was a potential inhibitor to δSHP-1, the catalytic domain of SHP-1 protein tyrosine phosphatase, and its IC(50) was 4ug/ml, and was more sensitive towards SHP-1than other PTPs, which indicated that SHP-1 might be a target of the extract of Perilla stem. It can strengthened the level of tyrosine phosphorylation of insulin receptor (IR) and extracellular signal-regulated protein kinase (ERK) in HepG2 cells, and then activated the insulin signaling pathway through inhibiting the protein phosphorylation of SHP-1. These results demonstrated that the extract of Perilla stem could play an important role for diabetes treatment through inhibiting the level of SHP-1 in insulin signaling pathway.


Subject(s)
Insulin/physiology , Perilla , Plant Extracts/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/antagonists & inhibitors , Signal Transduction/drug effects , Hep G2 Cells , Humans , Phosphorylation , Plant Stems
18.
Pain ; 156(4): 597-608, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25790452

ABSTRACT

Transient receptor potential vanilloid 1 (TRPV1) receptors are expressed in nociceptive neurons of rat dorsal root ganglions (DRGs) and mediate inflammatory pain. Nonspecific inhibition of protein-tyrosine phosphatases (PTPs) increases the tyrosine phosphorylation of TRPV1 and sensitizes TRPV1. However, less is known about tyrosine phosphorylation's implication in inflammatory pain, compared with that of serine/threonine phosphorylation. Src homology 2 domain-containing tyrosine phosphatase 1 (Shp-1) is a key phosphatase dephosphorylating TRPV1. In this study, we reported that Shp-1 colocalized with and bound to TRPV1 in nociceptive DRG neurons. Shp-1 inhibitors, including sodium stibogluconate and PTP inhibitor III, sensitized TRPV1 in cultured DRG neurons. In naive rats, intrathecal injection of Shp-1 inhibitors increased both TRPV1 and tyrosine-phosphorylated TRPV1 in DRGs and induced thermal hyperalgesia, which was abolished by pretreatment with TRPV1 antagonists capsazepine, BCTC, or AMG9810. Complete Freund's adjuvant (CFA)-induced inflammatory pain in rats significantly increased the expression of Shp-1, TRPV1, and tyrosine-phosphorylated TRPV1, as well as the colocalization of Shp-1 and TRPV1 in DRGs. Intrathecal injection of sodium stibogluconate aggravated CFA-induced inflammatory pain, whereas Shp-1 overexpression in DRG neurons alleviated it. These results suggested that Shp-1 dephosphorylated and inhibited TRPV1 in DRG neurons, contributing to maintain thermal nociceptive thresholds in normal rats, and as a compensatory mechanism, Shp-1 increased in DRGs of rats with CFA-induced inflammatory pain, which was involved in protecting against excessive thermal hyperalgesia.


Subject(s)
Enzyme Inhibitors/therapeutic use , Ganglia, Spinal/pathology , Neurons/drug effects , Pain/drug therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/therapeutic use , TRPV Cation Channels/metabolism , Animals , Calcium/metabolism , Capsaicin/pharmacology , Cell Culture Techniques , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Freund's Adjuvant/toxicity , Gene Expression Regulation/drug effects , Inflammation/chemically induced , Inflammation/complications , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/metabolism , Pain/etiology , Pain/pathology , Pain Threshold/drug effects , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
19.
World J Gastroenterol ; 20(41): 15269-74, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25386075

ABSTRACT

Hepatocellular carcinoma is one of the most common cancers worldwide, and a leading cause of cancer-related death. Owing to unsatisfactory clinical outcomes under the current standard of care, there is a need to search for and identify novel and potent therapeutic targets to improve patient outcomes. Sorafenib is the first and only approved targeted therapy for the treatment of hepatocellular carcinoma. Besides functioning as a multiple tyrosine kinase, sorafenib also acts via a kinase-independent mechanism to target signal transducer and activator of transcription 3 (STAT3) signaling in hepatocellular carcinoma cells. STAT3 is a key regulator of inflammation, cell survival, and tumorigenesis of liver cells, and the high percentage of hepatocellular carcinoma cells with constitutively active STAT3 justifies targeting it for the development of novel therapeutics. Sorafenib inactivates STAT3 and STAT3-related signaling by inducing a conformational change in and releasing the autoinhibition of Src homology region 2 domain-containing phosphatase-1. This phosphatase negatively regulates STAT3 activity, which leads to the subsequent apoptosis of cancer cells. The novel anti-cancer property of sorafenib will be discussed in this review, not only adding information regarding its mechanism of action but also providing an innovative approach for the development of cancer therapeutics in the future.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Protein Kinase Inhibitors/therapeutic use , STAT3 Transcription Factor/metabolism , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Down-Regulation , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Molecular Targeted Therapy , Niacinamide/therapeutic use , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Signal Transduction/drug effects , Sorafenib , Treatment Outcome
20.
Breast Cancer Res Treat ; 148(2): 279-89, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25301089

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) is implicated breast cancer metastasis and represents a potential target for developing new anti-tumor metastasis drugs. The purpose of this study is to investigate whether the natural agent 1'-acetoxychavicol acetate (ACA), derived from the rhizomes and seeds of Languas galanga, could suppress breast cancer metastasis by targeting STAT3 signaling pathway. ACA was examined for its effects on breast cancer migration/invasion and metastasis using Transwell assays in vitro and breast cancer skeletal metastasis mouse model in vivo (n = 10 mice per group). The inhibitory effect of ACA on cellular STAT3 signaling pathway was investigated by series of biochemistry analysis. The chavicol preferentially suppressed cancer cell migration and invasion, and this activity was superior to its cytotoxic effects. ACA suppressed both constitutive and interleukin-6-inducible STAT3 activation and diminished the accumulation of STAT3 in the nucleus and its DNA-binding activity. More importantly, ACA treatment led to significant up-regulation of Src homology region 2 domain-containing phosphatase 1 (SHP-1), and the ACA-induced depression of cancer cell migration and STAT3 signaling could be apparently reversed by blockade of SHP-1. Matrix metalloproteinase (MMP)-2 and -9, gene products of STAT3 that regulate cell invasion, were specifically suppressed by ACA. In tumor metastasis model, ACA potently inhibited the human breast cancer cell-induced osteolysis, and had little apparent in vivo toxicity at the test concentrations. ACA is a novel drug candidate for the inhibition of tumor metastasis through interference with the SHP-1/STAT3/MMPs signaling pathway.


Subject(s)
Benzyl Alcohols/therapeutic use , Bone Neoplasms/prevention & control , Breast Neoplasms/prevention & control , Matrix Metalloproteinase 3/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , STAT3 Transcription Factor/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Female , Fluorescent Antibody Technique , Humans , Matrix Metalloproteinase 3/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation/drug effects , Plant Extracts/therapeutic use , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL