Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Eur J Pharmacol ; 973: 176562, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38588767

ABSTRACT

In recent years, immunosuppressants have shown significant success in the treatment of autoimmune diseases. Therefore, there is an urgent need to develop additional immunosuppressants that offer more options for patients. Toosendanin has been shown to have immunosuppressive activity in vitro as well as effects on autoimmune hepatitis (AIH) in vivo. Toosendanin did not induce apoptosis in activated T-cells and affect the survival rate of naive T-cells. Toosendanin did not affect the expression of CD25 or secretion of IL-2 by activated T-cells, and not affect the expression of IL-4 and INF-γ. Toosendanin did not affect the phosphorylation of STAT5, ERK, AKT, P70S6K. However, toosendanin inhibited proliferation of anti-CD3/anti-CD28 mAbs-activated T-cells with IC50 of (10 ± 2.02) nM. Toosendanin arrested the cell cycle in the G0/G1 phase, significantly inhibited IL-6 and IL-17A secretion, promoted IL-10 expression, and inhibited the P38 MAPK pathway. Finally, toosendanin significantly alleviated ConA-induced AIH in mice. In Summary, toosendanin exhibited immunosuppressive activity in vivo and in vitro. Toosendanin inhibits the proliferation of activated T-cells through the P38 MAPK signalling pathway, significantly suppresses the expression of inflammatory factors, enhances the expression of anti-inflammatory factors, and effectively alleviates ConA-induced AIH in mice, suggesting that toosendanin may be a lead compound for the development of novel immunomodulatory agents with improved efficacy and reduced toxicity.


Subject(s)
Cell Proliferation , Drugs, Chinese Herbal , T-Lymphocytes , Triterpenes , p38 Mitogen-Activated Protein Kinases , Animals , Cell Proliferation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Mice , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Drugs, Chinese Herbal/pharmacology , MAP Kinase Signaling System/drug effects , Lymphocyte Activation/drug effects , Hepatitis, Autoimmune/drug therapy , Hepatitis, Autoimmune/immunology , Hepatitis, Autoimmune/pathology , Cytokines/metabolism , Immunosuppressive Agents/pharmacology , Mice, Inbred BALB C , Female
2.
Eur J Pharmacol ; 929: 175151, 2022 Aug 15.
Article in English | MEDLINE | ID: mdl-35841942

ABSTRACT

A series of novel scopariusicide derivatives were designed and synthesized starting from the main diterpenoid from the aerial parts of Isodon scoparius. Sis-25 was the most effective compound among them. The potential mechanism(s) of its immunosuppressive activity in vitro, as well as its effects on delayed type hypersensitivity (DTH) reaction and imiquimod-induced dermatitis in vivo were investigated in this study. Sis-25 inhibited anti-CD3/anti-CD28 mAbs, PHA or alloantigen-induced T cell proliferation without obvious cytotoxicity. Sis-25 was a highly selective inhibitor of GSK3-ß and inhibited the mTOR/p70S6K pathway but not the PI3K/Akt, p38 MAPK/ERK 1/2 and JAK3/STAT5 pathways. Furthermore, Sis-25 significantly inhibited IFN-γ, IL-6 and IL-17 expression but not IL-10 expression in activated T cells. Finally, Sis-25 treatment mitigated the DNFB-induced DTH reaction and ameliorated imiquimod-induced dermatitis. In summary, Sis-25 exerted significant immunosuppressive activity by targeting GSK3ß in vitro and in vivo. Sis-25 may guide the design of new drugs for more effective and safer treatments of autoimmune diseases and provide new insight into developing utilizations of Isodon scoparius.


Subject(s)
Cyclobutanes , Dermatitis , Cell Proliferation , Cyclobutanes/pharmacology , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinase 3 beta , Humans , Imiquimod , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism
5.
Oncol Lett ; 21(5): 390, 2021 May.
Article in English | MEDLINE | ID: mdl-33777213

ABSTRACT

Nicotinamide phosphoribosyltransferase (NAMPT) is a critical rate-limiting enzyme involved in NAD synthesis that has been shown to contribute to the progression of liver cancer. However, the potential role and mechanism of NAMPT in hepatitis B virus (HBV)-associated liver cancer remain unclear. The present study assessed the expression of NAMPT in HBV-positive and -negative liver cancer cells, and investigated whether HBV-induced NAMPT expression is dependent on HBV X protein (HBx). In addition, the role of NAMPT in HBV replication and transcription, and in HBV-mediated liver cancer cell growth was explored. The effects of NAMPT on the glycolytic pathway were also evaluated. Reverse transcription-quantitative PCR and western blotting results revealed that NAMPT expression levels were significantly higher in HBV-positive liver cancer cells than in HBV-negative liver cancer cells, and this effect was HBx-dependent. Moreover, the activation of NAMPT was demonstrated to be required for HBV replication and transcription. The NAMPT inhibitor FK866 repressed cell survival and promoted cell death in HBV-expressing liver cancer cells, and these effects were attenuated by nicotinamide mononucleotide. Furthermore, the inhibition of NAMPT was associated with decreased glucose uptake, decreased lactate production and decreased ATP levels in HBV-expressing liver cancer cells, indicating that NAMPT may promote the aerobic glycolysis. Collectively, these findings reveal a positive feedback loop in which HBV enhances NAMPT expression and the activation of NAMPT promotes HBV replication and HBV-mediated malignant cell growth in liver cancer. The present study highlights the important role of NAMPT in the regulation of aerobic glycolysis in HBV-mediated liver cancer, and suggests that NAMPT may be a promising treatment target for patients with HBV-associated liver cancer.

6.
Br J Cancer ; 123(7): 1154-1163, 2020 09.
Article in English | MEDLINE | ID: mdl-32632148

ABSTRACT

BACKGROUND: Hepatitis B virus (HBV) has a crucial role in the progression of hepatocellular carcinoma (HCC). Tumour cells must develop anoikis resistance in order to survive before metastasis. This study aimed to investigate the mechanism of IQGAP1 in HBV-mediated anoikis evasion and metastasis in HCC cells. METHODS: IQGAP1 expression was detected by immunohistochemistry, real-time PCR and immunoblot analysis. Lentiviral-mediated stable upregulation or knockdown of IGAQP1, immunoprecipitation, etc. were used in function and mechanism study. RESULTS: IQGAP1 was markedly upregulated in HBV-positive compared with HBV-negative HCC cells and tissues. IQGAP1 was positively correlated to poor prognosis of HBV-associated HCC patients. IQGAP1 overexpression significantly enhanced the anchorage-independent growth and metastasis, whereas IQGAP1-deficient HCC cells are more sensitive to anoikis. Mechanistically, we found that HBV-induced ROS enhanced the association of IQGAP1 and Rac1 that activated Rac1, leading to phosphorylation of Src/FAK pathway. Antioxidants efficiently inhibited IQGAP1-mediated anoikis resistance and metastasis. CONCLUSIONS: Our study indicated an important mechanism by which upregulated IQGAP1 by HBV promoted anoikis resistance, migration and invasion of HCC cells through Rac1-dependent ROS accumulation and activation of Src/FAK signalling, suggesting IQGAP1 as a prognostic indicator and a novel therapeutic target in HCC patients with HBV infection.


Subject(s)
Carcinoma, Hepatocellular/pathology , Focal Adhesion Kinase 1/physiology , Liver Neoplasms/pathology , Reactive Oxygen Species/metabolism , rac1 GTP-Binding Protein/physiology , ras GTPase-Activating Proteins/physiology , src-Family Kinases/physiology , Animals , Anoikis , Cell Line, Tumor , Female , Hepatitis B/complications , Humans , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Signal Transduction/physiology
7.
J Innate Immun ; 12(4): 304-320, 2020.
Article in English | MEDLINE | ID: mdl-31865314

ABSTRACT

Toll-like receptor 3 (TLR3) is a sensor of endogenous cell necrosis during the process of acute inflammation. Interleukin (IL)-1 receptor antagonist (IL-1Ra) is an anti-inflammatory cytokine and can negatively regulate the pathogenesis of inflammation. However, whether and how activation of TLR3 can regulate IL-1Ra expression has not been clarified. Here, we show that poly(I:C) induces IL-1Ra expression in primarily cultured human fibroblast-like synoviocytes and other types of cells. Induction of IL-1Ra by poly(I:C) was dependent on TLR3, but was independent of melanoma differentiation--associated protein 5 or retinoic acid-inducible gene I. Interferon regulatory factor 3 (IRF3) directly binds to the IL-1Ra promoter and promotes IL-1Ra expression in response to poly(I:C) stimulation. Induction of IL-1Ra by poly(I:C) was abolished by the inhibition of the NF-κB signaling, attenuated by the inhibition of the PI3K-Akt signaling, enhanced by inhibition of the ERK1/2 or MSK1/2 activation, but was independent of the p38 MAPK signaling. Treatment with poly(I:C) or Sendai virus elevated the levels of serum IL-1Ra in wild-type, but not in TLR3-/- or IRF3-/- mice. Our findings may provide new insights into the intrinsic anti-inflammatory function of TLR3 and double-stranded RNA-induced IL-Ra expression by TLR3 and its regulation.


Subject(s)
Interleukin 1 Receptor Antagonist Protein/immunology , Toll-Like Receptor 3/immunology , Animals , Cell Line , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Interferon Regulatory Factor-3/genetics , Interleukin 1 Receptor Antagonist Protein/genetics , Mice , Mice, Knockout , Poly I-C/pharmacology , Toll-Like Receptor 3/genetics
8.
Br J Pharmacol ; 177(7): 1666-1676, 2020 04.
Article in English | MEDLINE | ID: mdl-31724152

ABSTRACT

BACKGROUND AND PURPOSE: Immunosuppressive drugs have shown great promise in treating autoimmune diseases in recent years. A series of novel oxazole derivatives were screened for their immunosuppressive activity. PO-322 [1H-indole-2,3-dione 3-(1,3-benzoxazol-2-ylhydrazone)] was identified as the most effective of these compounds. Here, we have investigated the mechanism(s) underlying the inhibition of T-cell proliferation in vitro by PO-322, as well as its effects on the delayed-type hypersensitivity (DTH) response and imiquimod-induced dermatitis in vivo. EXPERIMENTAL APPROACH: T-cell proliferation and apoptosis were analysed with flow cytometry. Cell viability was assessed with a CCK-8 assay. Protein kinase activity was assessed by SelectScreen Kinase Profiling Services. The phosphorylation of signal-regulated molecules was measured by Western blot. Cytokine levels were determined by elisa. The effect of PO-322 on DTH and imiquimod-induced dermatitis was evaluated in BALB/c mice. KEY RESULTS: PO-322 inhibited human T-cell proliferation with anti-CD3/anti-CD28 mAbs or alloantigen without significant cytotoxicity. Importantly, PO-322 was a selective inhibitor of the serum- and glucocorticoid-regulated kinase 1 (SGK1) and decreased NDRG1 phosphorylation but not p70S6K, STAT5, Akt, or ERK1/2 phosphorylation. Furthermore, PO-322 inhibited IFN-γ, IL-6, and IL-17 expression but not IL-10 expression. Finally, treatment with PO-322 was safe and effective for ameliorating the DTH response and imiquimod-induced dermatitis in mice. CONCLUSIONS AND IMPLICATIONS: PO-322 exerted immunosuppressive activity in vitro and in vivo by selectively inhibiting SGK1 activity. PO-322 represents a potential lead compound for the design and development of new drugs for the treatment of autoimmune diseases.


Subject(s)
Lymphocyte Activation , T-Lymphocytes , Animals , Cell Proliferation , Cytokines , Immunosuppressive Agents , Mice , Mice, Inbred BALB C
9.
Front Pharmacol ; 10: 1579, 2019.
Article in English | MEDLINE | ID: mdl-32063843

ABSTRACT

As a GSK-3ß inhibitor reported by our group, K313 is a novel benzoxazole derivative and displays anti-inflammatory properties in RAW264.7 macrophages without cytotoxicity. The activity of GSK-3ß affects the differentiation and maturation of bone marrow-derived dendritic cells (DCs). This study aims to investigate whether K313 can be used to induce regulatory/tolerogenic dendritic cells (DCregs), and the therapeutic effects of DCregs induced by K313 in the autoimmune model of experimental autoimmune encephalitis (EAE). The results show that compared with LPS stimulated mature DCs, K313-treated bone marrow-derived DCs display obvious tolerogenic characteristics with decreased expression of co-stimulatory molecules, downregulated secretions of pro-inflammatory cytokines and unregulated secretion of anti-inflammatory cytokine IL-10. The above characteristics conform to the typical phenotypes of DCregs. Moreover, K313-modified DCregs inhibit antigen-specific T cell responses in vitro. Furthermore, by adoptive transfer, K313 modified DCregs to the EAE mice, and the development of disease was ameliorated to some extent. In addition, treatment with K313-modified DCregs also significantly reduced the percentages of splenetic Th1 and Th17 cells and increased the percentage of regulatory T cells in EAE mice. In conclusion, K313-modified DCregs show anti-inflammatory properties in vitro and have a significant positive effect on the EAE disease in vivo. Our data indicate that K313-induced DCregs pulsed with auto-antigen might have potential use as a therapeutic approach for autoimmune inflammation of the central nervous system.

10.
J Cell Biochem ; 120(6): 9193-9202, 2019 06.
Article in English | MEDLINE | ID: mdl-30506723

ABSTRACT

Immunosuppressants have shown striking achievements in treating autoimmune diseases in recent years. It is urgent to develop more immunosuppressants to provide more options for patients. PO-296 [2-(6-chlorobenzo[d]oxazol-2-yl)-4,5,6,7-tetrahydro-2H-indazol-3-ol] was identified as a novel benzoxazole derivative. We observed that it exhibits an obvious immunosuppressive activity to T lymphocytes. PO-296 significantly inhibited the proliferation of activated human T lymphocyte without cytotoxicity. Moreover, PO-296 did not affect the expression of cluster of differentiation (CD)-25 or CD69 but induced T lymphocyte cycle arrest in the G0/G1 phase. Furthermore, PO-296 inhibited interleukin (IL)-6, IL-17, and interferon gamma expression but had no effect on IL-2, IL-4, or IL-10. Yet, importantly, PO-296 inhibited the phosphorylation of signal transducer and activator of transcription 5 (STAT5), increased the phosphorylation of p70S6K, but did not affect the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mitogen-activated protein kinase pathway. In conclusion, these findings indicate that PO-296 inhibits human activated T-lymphocyte proliferation by affecting the janus kinase 3 (JAK3)/STAT5 pathway. PO-296 possesses a potential lead compound for the design and development of new immunosuppressants for the treatment of autoimmune diseases.


Subject(s)
Benzoxazoles/chemistry , Benzoxazoles/pharmacology , Lymphocyte Activation/drug effects , Phosphatidylinositol 3-Kinases/metabolism , STAT5 Transcription Factor/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Survival/drug effects , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-17/metabolism , Interleukin-2/metabolism , Interleukin-4/metabolism , Interleukin-6/metabolism , T-Lymphocytes/cytology
11.
J Cell Biochem ; 119(7): 5382-5390, 2018 07.
Article in English | MEDLINE | ID: mdl-29345351

ABSTRACT

Benzoxazole and its derivatives have been widely studied in recent years due to their various biological properties. A previous study has demonstrated that K313 (1H-indole-2,3-dione 3-(1,3-benzoxazol-2-ylhydrazone)), a novel benzoxazole derivative, inhibits T cell proliferation to yield immunosuppressive effects. However, there are no related reports about its anti-inflammatory effects. In the present study, we investigated the anti-inflammatory properties and the underlying molecular mechanism of K313 in lipopolysaccharide (LPS)-induced RAW264.7 macrophages. K313 dose-dependently (5, 10, and 20 µM) inhibited LPS-stimulated nitric oxide (NO), interleukin (IL)-6, tumor necrosis factor (TNF)-α, and 3-nitrotyrosine (3-NT) production and significantly decreased the gene transcription levels of inducible nitric oxide (iNOS), IL-6, and TNF-α. In addition, the results showed that the inflammatory cytokines suppressed by K313 were not regulated by p65 NF-κB, ERK1/2, AKT, or p38 MAPK. Instead, K313 increased phosphorylation of glycogen synthase kinase-3 beta (GSK-3ß) (Ser9) resulting in GSK-3ß deactivation. Moreover, in LPS-stimulated RAW264.7 macrophages, K313 and lithium chloride (LiCl) had a synergistic effect on the anti-inflammatory response. These results indicated that K313 exhibited anti-inflammatory properties and revealed the potential mechanism. K313 can increase GSK-3ß (Ser9) phosphorylation to decrease GSK-3ß activation in LPS-induced RAW264.7 macrophages.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzoxazoles/pharmacology , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Inflammation/drug therapy , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Benzoxazoles/chemistry , Cells, Cultured , Cytokines/metabolism , Inflammation/immunology , Inflammation/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Int J Oncol ; 49(5): 2127-2134, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27600372

ABSTRACT

Benzothiazole derivatives are known for various biological activities, and their potency in cancer therapy have received considerable attention in recent years. However, the poor water solubility of most benzothiazole derivatives has limited their clinical application. We developed BD926, a novel water-soluble benzothiazole derivative and showed here that it could inhibit the proliferation and induce apoptosis of human Ramos B-lymphoma cells. We further showed that BD926 triggered apoptosis through both mitochondria and endoplasmic reticulum pathways. Moreover, BD926 caused cell cycle arrest at G0/G1 stage. Furthermore, accumulation of reactive oxygen species (ROS) were observed after BD926 treatment and ROS inhibitor was able to attenuate BD926-induced apoptosis, which suggested that BD926-induced apoptosis may be due to over-producing ROS. These results demonstrate the anticancer effects of BD926 in cell models and raise the possibility for the application of BD926 in cancer therapy.


Subject(s)
Apoptosis/drug effects , B-Lymphocytes/pathology , Benzothiazoles/pharmacology , Endoplasmic Reticulum/drug effects , Lymphoma, B-Cell/pathology , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Benzothiazoles/chemistry , Blotting, Western , Endoplasmic Reticulum/metabolism , Flow Cytometry , Humans , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tumor Cells, Cultured
13.
J Immunol Res ; 2016: 5021537, 2016.
Article in English | MEDLINE | ID: mdl-28070525

ABSTRACT

It is well known that dendritic cells (DCs) play a pivotal role in triggering self-specific responses. Conversely, tolerogenic DCs (tolDCs), a specialized subset, induce tolerance and negatively regulate autoreactive responses. Tofacitinib, a Janus kinase inhibitor developed by Pfizer for treatment of rheumatoid arthritis, is probable to be a promising candidate for inducing tolDCs. The aims of this study were to evaluate the effectiveness of tolDCs induced by tofacitinib in a myelin oligodendrocyte glycoprotein- (MOG-) specific experimental autoimmune encephalomyelitis (EAE) model and to investigate their effects on Th17/Treg balance in the animal model of multiple sclerosis (MS). Our results revealed that tofacitinib-treated DCs maintained a steady semimature phenotype with a low level of proinflammatory cytokines and costimulatory molecules. DCs treated by tofacitinib also induced antigen-specific T cells hyporesponsiveness in a concentration-dependent manner. Upon intravenous injection into EAE mice, MOG pulsed tolDCs significantly dampened disease activity, and adoptive cell therapy (ACT) disturbed Th17/Treg balance with a remarkable decrease of Th1/Th17 cells and an increase in regulatory T cells (Tregs). Overall, DCs modified by tofacitinib exhibited a typical tolerogenic phenotype, and the antigen-specific tolDCs may represent a new avenue of research for the development of future clinical treatments for MS.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/transplantation , Encephalomyelitis, Autoimmune, Experimental/therapy , Immune Tolerance/immunology , Immunotherapy, Adoptive , Piperidines/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , T-Lymphocytes, Regulatory/cytology , Th17 Cells/cytology , Animals , CD4 Lymphocyte Count , Cells, Cultured , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Protein Kinase Inhibitors/pharmacology , Th1 Cells/cytology
14.
Eur J Pharmacol ; 761: 36-43, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25935419

ABSTRACT

Immunosuppressants are widely used for treatment of T cell-mediated autoimmune diseases and allogeneic graft rejection. However, because of the toxicity and tolerance of these drugs, novel immunosuppressants are urgently needed. We synthesized a series of novel water-soluble benzothiazole derivatives and found that BD926 [sodium 2-(benzo[d]thiazol-2-yl)-4,5,6,7-tetrahydro-2H-indazol-3-olate] had potent immunosuppressive activity. Treatment with BD926 significantly inhibited anti-CD3/anti-CD28 and alloantigen-induced human T cell proliferation as well as IL2-stimulated activated T cell proliferation in a dose-dependent manner in vitro. BD926 had no obvious cytotoxicity against human resting T cells, IL-4 treated activated T cells and fibroblast-like synoviocytes in our experimental conditions. Furthermore, BD926 induced cell cycle arrest at G0/G1 phase and inhibited the cyclin D3 and CDK 6 expression in activated T cells. BD926 inhibited the STAT5, but not Akt and p70S6K, phosphorylation in a dose-dependent manner in the IL-2-treated activated T cells. Interestingly, BD926 inhibited IFN-γ, IL-6 and IL-17, but not IL-2, IL-4 and IL-10, production in activated T cells. Finally, treatment with BD926 reduced delayed-type hypersensitivity in mice in a dose-dependent manner. Collectively, these data suggest that BD926 may be a lead compound for the design and development of new immunosuppressants for the intervention of allograft rejection and autoimmune diseases.


Subject(s)
Benzimidazoles/pharmacology , Benzothiazoles/pharmacology , Cell Proliferation/drug effects , Immunosuppressive Agents/pharmacology , Indazoles/pharmacology , Lymphocyte Activation/drug effects , STAT5 Transcription Factor/metabolism , T-Lymphocytes/drug effects , Water/chemistry , Animals , Benzimidazoles/chemistry , Benzothiazoles/chemistry , Cells, Cultured , Cyclin D3/metabolism , Cyclin-Dependent Kinase 6/metabolism , Cytokines/metabolism , Dermatitis, Allergic Contact/immunology , Dermatitis, Allergic Contact/metabolism , Dermatitis, Allergic Contact/prevention & control , Dinitrofluorobenzene , Disease Models, Animal , Dose-Response Relationship, Drug , Down-Regulation , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Immunosuppressive Agents/chemistry , Indazoles/chemistry , Mice, Inbred BALB C , Phosphorylation , Signal Transduction/drug effects , Solubility , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
15.
Molecules ; 19(11): 17173-86, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25347460

ABSTRACT

In this study, a benzimidazole derivative named BMT-1 is revealed as a potential immunomodulatory agent. BMT-1 inhibits the activity of H+/K+-ATPases from anti-CD3/CD28 activated T cells. Furthermore, inhibition the H+/K+-ATPases by use of BMT-1 should lead to intracellular acidification, inhibiting T cell proliferation. To explore this possibility, the effect of BMT-1 on intracellular pH changes was examined by using BCECF as a pH-dependent fluorescent dye. Interestingly, increases in the pHi were observed in activated T cells, and T cells treated with BMT-1 showed a more acidic intracellular pH. Finally, BMT-1 targeted the H+/K+-ATPases and inhibited the proliferative response of anti-CD3/CD28-stimulated T cells. A cell cycle analysis indicated that BMT-1 arrested the cell cycle progression of activated T cells from the G1 to the S phase without affecting CD25 expression or interleukin-2 (IL-2) production; treating IL-2-dependent PBMCs with BMT-1 also led to the inhibition of cell proliferation. Taken together, these findings demonstrate that BMT-1 inhibits the proliferation of T cells by interfering with H+/K+-ATPases and down-regulating intracellular pHi. This molecule may be an interesting lead compound for the development of new immunomodulatory agents.


Subject(s)
Benzimidazoles/pharmacology , Cell Proliferation/drug effects , H(+)-K(+)-Exchanging ATPase/metabolism , Proton Pump Inhibitors/pharmacology , T-Lymphocytes/drug effects , CD28 Antigens/metabolism , CD3 Complex/metabolism , Cell Cycle Checkpoints/drug effects , Cells, Cultured , Down-Regulation/drug effects , Humans , Hydrogen-Ion Concentration , Immunologic Factors/pharmacology , Interleukin-2/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/drug effects , T-Lymphocytes/metabolism
16.
Molecules ; 17(2): 2015-29, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22343406

ABSTRACT

Human mitotic kinesin Eg5 plays an essential role in mitoses and is an interesting drug target against cancer. To find the correlation between Eg5 and its inhibitors, structure-based 3D-quantitative structure-activity relationship (QSAR) studies were performed on a series of dihydropyrazole and dihydropyrrole derivatives using comparative molecular field analysis (CoMFA) and comparative molecular similarity indices analysis (CoMSIA) methods. Based on the LigandFit docking results, predictive 3D-QSAR models were established, with cross-validated coefficient values (q²) up to 0.798 for CoMFA and 0.848 for CoMSIA, respectively. Furthermore, the CoMFA and CoMSIA models were mapped back to the binding sites of Eg5, which could provide a better understanding of vital interactions between the inhibitors and the kinase. Ligands binding in hydrophobic part of the inhibitor-binding pocket were found to be crucial for potent ligand binding and kinases selectivity. The analyses may be used to design more potent EG5 inhibitors and predict their activities prior to synthesis.


Subject(s)
Kinesins/antagonists & inhibitors , Kinesins/chemistry , Mitosis/drug effects , Pyrroles/chemistry , Pyrroles/pharmacology , Binding Sites , Humans , Ligands , Models, Molecular , Protein Binding , Quantitative Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...