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










Publication year range
1.
Theranostics ; 11(15): 7527-7545, 2021.
Article in English | MEDLINE | ID: mdl-34158865

ABSTRACT

Rationale: One of the most common metabolic defects in cancers is the deficiency in arginine synthesis, which has been exploited therapeutically. Yet, challenges remain, and the mechanisms of arginine-starvation induced killing are largely unclear. Here, we sought to demonstrate the underlying mechanisms by which arginine starvation-induced cell death and to develop a dietary arginine-restriction xenograft model to study the in vivo effects. Methods: Multiple castration-resistant prostate cancer cell lines were treated with arginine starvation followed by comprehensive analysis of microarray, RNA-seq and ChIP-seq were to identify the molecular and epigenetic pathways affected by arginine starvation. Metabolomics and Seahorse Flux analyses were used to determine the metabolic profiles. A dietary arginine-restriction xenograft mouse model was developed to assess the effects of arginine starvation on tumor growth and inflammatory responses. Results: We showed that arginine starvation coordinately and epigenetically suppressed gene expressions, including those involved in oxidative phosphorylation and DNA repair, resulting in DNA damage, chromatin-leakage and cGAS-STING activation, accompanied by the upregulation of type I interferon response. We further demonstrated that arginine starvation-caused depletion of α-ketoglutarate and inactivation of histone demethylases are the underlying causes of epigenetic silencing. Significantly, our dietary arginine-restriction model showed that arginine starvation suppressed prostate cancer growth in vivo, with evidence of enhanced interferon responses and recruitment of immune cells. Conclusions: Arginine-starvation induces tumor cell killing by metabolite depletion and epigenetic silencing of metabolic genes, leading to DNA damage and chromatin leakage. The resulting cGAS-STING activation may further enhance these killing effects.


Subject(s)
Arginine/deficiency , Chromatin/metabolism , DNA Repair , Gene Expression Regulation, Neoplastic , Gene Silencing , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Nucleotidyltransferases/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Chromatin/genetics , Chromatin/pathology , Humans , Male , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Nucleotidyltransferases/genetics , PC-3 Cells , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology
2.
Mater Sci Eng C Mater Biol Appl ; 100: 308-314, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30948066

ABSTRACT

In this study, we used electrospinning to prepare a bilayered polycaprolactone (PCL) tubular graft consisting of an internal layer comprising axial nanofibers and an external layer comprising circumferentially aligned nanofibers. Subsequently, the surfaces of the electrospun PCL tubular scaffolds were modified with 1,6-diaminohexane to introduce amino groups and were then chemically conjugated with gelatin (Gel). The amino groups and Gel were successfully immobilized on the PCL scaffolds according to a ninhydrin assay, attenuated total reflectance Fourier transform infrared (ATR-FTIR) spectroscopic analysis and contact angle analysis. Additionally, vascular smooth muscle cells (vSMCs, A7r5) were cultured on random and aligned Gel-PCL scaffolds to evaluate the effects of fiber orientation on cell behavior. The results of immunofluorescence analysis showed that vSMCs on the aligned Gel-PCL scaffolds exhibited a pro-contractile phenotype.


Subject(s)
Myocytes, Smooth Muscle/cytology , Polyesters/pharmacology , Tissue Scaffolds/chemistry , Amines/chemistry , Animals , Cell Shape/drug effects , Cells, Cultured , Immobilized Proteins/metabolism , Myocytes, Smooth Muscle/drug effects , Rats
3.
Front Immunol ; 8: 1424, 2017.
Article in English | MEDLINE | ID: mdl-29163499

ABSTRACT

The inhibitory effect of immunoreceptor tyrosine-based activation motif (ITAM)-containing adapters DAP12 and FcεRI γ-chain (FcRγ) has been found in many immune functions. Herein, we have further explored the role of these adapters in C-type lectin receptors response. We identified that FcRγ, but not DAP12, could negatively regulate the Dectin-1 responses in dendritic cells (DCs). Loss of FcRγ or both DAP12 and FcRγ enhanced the maturation and cytokine production in DCs upon Dectin-1 activation compared to normal cells, whereas DCs lacking only DAP12 showed little changes. In addition, increments of T cell activation and T helper 17 polarization induced by FcRγ-deficient DCs were observed both in vitro and in vivo. Examining the Dectin-1 signaling, we revealed that the activations of several signaling molecules were augmented in FcRγ-deficient DCs stimulated with Dectin-1 ligands. Furthermore, we demonstrated that the association of phosphatases SHP-1 and PTEN with FcRγ may contribute to the negative regulation of FcRγ in Dectin-1 activation in DCs. These results extend the inhibitory effect of ITAM-containing adapters to Dectin-1 response in immune functions, even though Dectin-1 contains an ITAM-like intracellular domain. According to the role of Dectin-1 in responding to microbes and tumor cells, our finding may have applications in the development of vaccine and cancer therapy.

4.
Nat Commun ; 8: 15874, 2017 06 22.
Article in English | MEDLINE | ID: mdl-28639619

ABSTRACT

Long noncoding RNAs (lncRNAs) have been implicated in hypoxia/HIF-1-associated cancer progression through largely unknown mechanisms. Here we identify MIR31HG as a hypoxia-inducible lncRNA and therefore we name it LncHIFCAR (long noncoding HIF-1α co-activating RNA); we describe its oncogenic role as a HIF-1α co-activator that regulates the HIF-1 transcriptional network, crucial for cancer development. Extensive analyses of clinical data indicate LncHIFCAR level is substantially upregulated in oral carcinoma, significantly associated with poor clinical outcomes and representing an independent prognostic predictor. Overexpression of LncHIFCAR induces pseudo-hypoxic gene signature, whereas knockdown of LncHIFCAR impairs the hypoxia-induced HIF-1α transactivation, sphere-forming ability, metabolic shift and metastatic potential in vitro and in vivo. Mechanistically, LncHIFCAR forms a complex with HIF-1α via direct binding and facilitates the recruitment of HIF-1α and p300 cofactor to the target promoters. Our results uncover an lncRNA-mediated mechanism for HIF-1 activation and establish the clinical values of LncHIFCAR in prognosis and potential therapeutic strategy for oral carcinoma.


Subject(s)
Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mouth Neoplasms/genetics , RNA, Long Noncoding/genetics , Animals , Biomarkers, Tumor/genetics , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice, Nude , Middle Aged , Mouth Neoplasms/mortality , Mouth Neoplasms/pathology , Prognosis , Proportional Hazards Models , Survival Analysis , Tumor Hypoxia/genetics , Xenograft Model Antitumor Assays
5.
Oncotarget ; 7(24): 36988-36999, 2016 Jun 14.
Article in English | MEDLINE | ID: mdl-27175590

ABSTRACT

14-3-3σ overexpression results in enhanced hepatocellular carcinoma (HCC) cell migration and HCC tumor vascular-invasion is significantly associated with 14-3-3σ expression. However, increased expression of 14-3-3σ paradoxically suppresses in vitro cell invasion of HCC. We hypothesize that surrounding tumor-associated stromal cells play a crucial role in 14-3-3σ-regulated HCC cell invasion. In this study, H68 fibroblasts, THP-1 and phorbol-12-myristate-13-acetate (PMA)-treated THP-1 (PMA-THP-1) cells were incubated with conditioned media of control (control-CM) and 14-3-3σ-overepxressing cells (14-3-3σ-CM), followed by co-culture with HCC cells. Invasiveness of HCC cells was examined by a Boyden chamber assay. HCC cells co-cultured with 14-3-3σ-CM treated cells significantly enhanced their invasive ability compared with control-CM treated cells. Moreover, incubation with 14-3-3σ-CM induced differential expression profiles of matrix metalloproteinases (MMPs) in fibroblasts (MMP-1, MMP-2, MMP-9, MMP-12 and MMP-14), THP-1 (MMP-1 and MMP-12) and PMA-THP-1 cells (MMP-2, MMP-12 and MMP-14). In contrast, silencing of 14-3-3σ by siRNA significantly abolished 14-3-3σ-CM induced MMPs. In addition, treatment with recombinant 14-3-3σ (r14-3-3σ) protein exhibits a similar expression profile of MMPs induced by 14-3-3σ-CM in fibroblasts, THP-1 and PMA-THP-1 cells. Finally, knockdown of aminopeptidase N (APN) significantly abrogated r14-3-3σ induced expression of MMPs in HS68 fibroblasts. These results suggest that HCC-secreted 14-3-3σ promotes expression of MMPs in cancerous surrounding cells via an APN dependent mechanism. 14-3-3σ has a paracrine effect in educating stromal cells in tumor-associated microenvironment.


Subject(s)
14-3-3 Proteins/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/pathology , Exoribonucleases/metabolism , Liver Neoplasms/pathology , Matrix Metalloproteinases/biosynthesis , Paracrine Communication/physiology , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Humans , Liver Neoplasms/metabolism , Neoplasm Invasiveness/pathology , Tumor Microenvironment/physiology
6.
Int J Cardiol ; 201: 441-8, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26313863

ABSTRACT

BACKGROUND: Rho-associated kinase (ROCK) plays an important role in maintaining embryonic stem (ES) cell pluripotency. To determine whether ROCK is involved in ES cell differentiation into cardiac and hematopoietic lineages, we evaluated the effect of ROCK inhibitors, Y-27632 and fasudil on murine ES and induced pluripotent stem (iPS) cell differentiation. METHODS: Gene expression levels were determined by real-time PCR, Western blot analysis and immunofluorescent confocal microscopy. Cell transplantation of induced differentiated cells were assessed in vivo in a mouse model (three groups, n=8/group) of acute myocardial infarction (MI). The cell engraftment was examined by immunohistochemical staining and the outcome was analyzed by echocardiography. RESULTS: Cells were cultured in hematopoietic differentiation medium in the presence or absence of ROCK inhibitor and colony formation as well as markers of ES, hematopoietic stem cells (HSC) and cells of cardiac lineages were analyzed. ROCK inhibition resulted in a drastic change in colony morphology accompanied by loss of hematopoietic markers (GATA-1, CD41 and ß-Major) and expressed markers of cardiac lineages (GATA-4, Isl-1, Tbx-5, Tbx-20, MLC-2a, MLC-2v, α-MHC, cTnI and cTnT) in murine ES and iPS cells. Fasudil-induced cardiac progenitor (Mesp-1 expressing) cells were infused into a murine MI model. They engrafted into the peri-infarct and infarct regions and preserved left ventricular function. CONCLUSIONS: These findings provide new insights into the signaling required for ES cell differentiation into hematopoietic as well as cardiac lineages and suggest that ROCK inhibitors are useful in directing iPS cell differentiation into cardiac progenitor cells for cell therapy of cardiovascular diseases.


Subject(s)
Embryonic Stem Cells/drug effects , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Pluripotent Stem Cells/drug effects , Protein Kinase Inhibitors/pharmacology , rho-Associated Kinases/antagonists & inhibitors , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Amides/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Cell Differentiation/drug effects , Echocardiography , Embryonic Stem Cells/cytology , Embryonic Stem Cells/enzymology , Gene Expression , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Male , Mice , Myocardial Infarction/therapy , Myocytes, Cardiac/cytology , Platelet Membrane Glycoprotein IIb/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/enzymology , Pyridines/pharmacology , Real-Time Polymerase Chain Reaction/methods , Stem Cell Transplantation/methods , rho-Associated Kinases/metabolism
7.
Proc Natl Acad Sci U S A ; 111(52): 18697-702, 2014 Dec 30.
Article in English | MEDLINE | ID: mdl-25512540

ABSTRACT

Long noncoding RNAs (lncRNAs) have been implicated in a variety of physiological and pathological processes, including cancer. In prostate cancer, prostate cancer gene expression marker 1 (PCGEM1) is an androgen-induced prostate-specific lncRNA whose overexpression is highly associated with prostate tumors. PCGEM1's tumorigenic potential has been recently shown to be in part due to its ability to activate androgen receptor (AR). Here, we report a novel function of PCGEM1 that provides growth advantages for cancer cells by regulating tumor metabolism via c-Myc activation. PCGEM1 promotes glucose uptake for aerobic glycolysis, coupling with the pentose phosphate shunt to facilitate biosynthesis of nucleotide and lipid, and generates NADPH for redox homeostasis. We show that PCGEM1 regulates metabolism at a transcriptional level that affects multiple metabolic pathways, including glucose and glutamine metabolism, the pentose phosphate pathway, nucleotide and fatty acid biosynthesis, and the tricarboxylic acid cycle. The PCGEM1-mediated gene regulation takes place in part through AR activation, but predominantly through c-Myc activation, regardless of hormone or AR status. Significantly, PCGEM1 binds directly to target promoters, physically interacts with c-Myc, promotes chromatin recruitment of c-Myc, and enhances its transactivation activity. We also identified a c-Myc binding domain on PCGEM1 that contributes to the PCGEM1-dependent c-Myc activation and target induction. Together, our data uncover PCGEM1 as a key transcriptional regulator of central metabolic pathways in prostate cancer cells. By being a coactivator for both c-Myc and AR, PCGEM1 reprograms the androgen network and the central metabolism in a tumor-specific way, making it a promising target for therapeutic intervention.


Subject(s)
Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA, Long Noncoding/metabolism , RNA, Neoplasm/metabolism , Aerobiosis/genetics , Cell Line, Tumor , Glycolysis/genetics , HEK293 Cells , Humans , Male , NADP/genetics , NADP/metabolism , Pentose Phosphate Pathway/genetics , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Structure, Tertiary , Proto-Oncogene Proteins c-myc/genetics , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics , Receptors, Androgen/genetics , Receptors, Androgen/metabolism
8.
PLoS One ; 7(7): e40873, 2012.
Article in English | MEDLINE | ID: mdl-22844415

ABSTRACT

We have previously demonstrated that the meningococcal antigen Ag473 in the presence of Freund's adjuvant can elicit protective immune responses in mouse challenge model. In this study, we evaluated the structural requirement for the immunological activity and the possible signaling pathway of recombinant Ag473 antigen produced in E. coli. We found that lipidated Ag473 (L-Ag473) possesses an intrinsic adjuvant activity that could be attributed to its ability to activate dendritic cells and promote their maturation. In addition, we found that L-Ag473 can activate human monocytes and promote maturation of human monocyte-derived dendritic cells. These results provide an indirect support that L-Ag473 may also be immunogenic in human. Interestingly, the observed activity is dependent on the overall conformation of L-Ag473 because heating and proteinase K treatment can diminish and abolish the activity. Furthermore, our data suggest a species-differential TLR recognition of L-Ag473. Overall, these data suggest a new paradigm for the ligand-TLR interaction in addition to demonstrating the self-adjuvanting activity of the vaccine candidate L-Ag473.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Immunomodulation , Lipids/chemistry , Lipoproteins/chemistry , Lipoproteins/immunology , Amino Acid Sequence , Animals , Bacterial Proteins/metabolism , Bone Marrow Cells/cytology , Cell Line , Chemokines/biosynthesis , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Lipoproteins/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Molecular Sequence Data , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Toll-Like Receptor 4/metabolism
9.
Immunobiology ; 216(10): 1103-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21621872

ABSTRACT

Dendritic cells (DCs) are the critical leukocytes in regulating immune responses. Accordingly, DCs are the major target in the development of immunomodulators. In this study, we examined the effect of Semen cuscutae (SC), an important traditional Chinese medicine, on mouse bone marrow-derived DCs. We found that the n-butanol and methanol extracts of SC significantly suppressed LPS-stimulated DC activation. Several flavonoids were verified in the extracts using HPLC, and then kaempferol was identified as the major flavonoid in the methanol fraction of SC. Kaempferol was able to reduce cytokines and chemokines produced by LPS-stimulated DCs, and this reduction was not due to its cytotoxicity on DCs. In addition, DC maturation was impaired by kaempferol. Furthermore, kaempferol abrogated the ability of LPS-stimulated DCs to promote Ag-specific T cell activation, both in vitro and in vivo. Thus, we show for the first time that SC exhibits an immunosuppressive effect on DCs and that the active ingredient kaempferol attenuates DC function, which suggests that kaempferol has potential in the treatment of chronic inflammatory and autoimmune diseases.


Subject(s)
Cuscuta/chemistry , Dendritic Cells/drug effects , Dendritic Cells/immunology , Drugs, Chinese Herbal/pharmacology , Kaempferols/pharmacology , Animals , Cell Aggregation/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Cytokines/biosynthesis , Dendritic Cells/cytology , Drugs, Chinese Herbal/toxicity , Kaempferols/toxicity , Lipopolysaccharides/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
10.
Cancer Immunol Immunother ; 60(7): 1019-27, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21499904

ABSTRACT

DNA vaccine has been suggested to use in cancer therapy, but the efficacy remains to be improved. The immunostimulatory effect of a fungal immunomodulatory protein Ling Zhi-8 (LZ-8) isolated from Ganoderma lucidum has been reported. In this study, we tested the adjuvanticity of LZ-8 for HER-2/neu DNA vaccine against p185(neu) expressing tumor MBT-2 in mice. We found that recombinant LZ-8 stimulated mouse bone marrow-derived dendritic cells (DCs) via TLR4 and its stimulatory effect was not due to any microbe contaminant. In addition, LZ-8 enhanced the ability of DCs to induce antigen-specific T cell activation in vitro and in a subunit vaccine model in vivo. Surprisingly, LZ-8 cotreatment strongly improved the therapeutic effect of DNA vaccine against MBT-2 tumor in mice. This increase in antitumor activity was attributed to the enhancement of vaccine-induced Th1 and CTL responses. Consistent with the results from DCs, the promoting effect of LZ-8 on DNA vaccine was diminished when the MBT-2 tumor cells were grown in TLR4 mutant mice. Thus, we concluded that LZ-8 may be a promising adjuvant to enhance the efficacy of DNA vaccine by activating DCs via TLR4.


Subject(s)
Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Fungal Proteins/immunology , Urinary Bladder Neoplasms/therapy , Vaccines, DNA/therapeutic use , Adjuvants, Immunologic , Animals , Blotting, Western , Disease Models, Animal , Fungal Proteins/genetics , Lymphocyte Activation , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , RNA, Messenger/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Cytotoxic/immunology , Transduction, Genetic , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology
11.
J Virol ; 84(22): 12011-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20844030

ABSTRACT

Since dendritic cells may play a key role in defense against influenza virus infection, we examined the effects of recombinant hemagglutinin (HA) proteins derived from mouse-adapted H1N1 (A/WSN/1933), swine-origin 2009 pandemic H1N1 (A/Texas/05/2009), and highly pathogenic avian influenza H5N1 (A/Thailand/KAN-1/2004) viruses on mouse myeloid dendritic cells (mDCs). The results reveal that tumor necrosis factor alpha (TNF-α), interleukin-12 (IL-12) p70, and major histocompatibility complex class II (MHC-II) expression was increased in mDCs after treatment with recombinant HA proteins of H1N1 and H5N1. The specificity of recombinant HA treatments for mDC activation was diminished after proteinase K digestion. HA apparently promotes mDC maturation by enhancing CD40 and CD86 expression and suppressing endocytosis. No significant differences in mDC activation were observed among recombinant proteins of H1N1 and H5N1. The stimulation of mDCs by HA proteins of H1N1 and H5N1 was completely MyD88 dependent. These findings may provide useful information for the development of more-effective influenza vaccines.


Subject(s)
Dendritic Cells/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza, Human/immunology , Animals , Cells, Cultured , Cytokines/immunology , Dendritic Cells/virology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/genetics , Influenza, Human/virology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Recombinant Proteins/genetics , Recombinant Proteins/immunology
12.
J Immunol ; 184(12): 6815-21, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20483746

ABSTRACT

Dendritic cells (DCs) play a crucial role in linking innate and adaptive immunity. Thus, DCs have been regarded as a major target of immunosuppressants for the control of harmful immune responses. In this study, we examined the effect of quercetin, a natural flavonoid found in many vegetables and fruits, on the activation and function of mouse DCs. Quercetin effectively inhibited LPS-induced DC activation by reducing the production of proinflammatory cytokines/chemokines and the expression levels of MHC class II and costimulatory molecules. In addition, quercetin uniquely blocked endocytosis by DCs and the LPS-induced DC migration was diminished by quercetin treatment. Furthermore, quercetin abrogated the ability of LPS-stimulated DCs to induce Ag-specific T cell activation, both in vitro and in vivo. Remarkably, coadministration of quercetin with 2,4-dinitro-1-fluorobenzene prevented 2,4-dinitro-1-fluorobenzene-induced contact hypersensitivity, indicating the potential of quercetin for treating delayed-type hypersensitive diseases. Blockage of LPS-induced ERK, JNK, Akt, and NF-kappaB activation contributed to the inhibitory effect of quercetin on DCs. These results strongly suggest that quercetin may be a potent immunosuppressive agent and could be used in the prevention and therapy of chronic inflammation, autoimmunity, and transplantation via the abolishment of DC activation and function.


Subject(s)
Dendritic Cells/drug effects , Immunosuppressive Agents/pharmacology , Quercetin/pharmacology , Animals , Blotting, Western , Chemotaxis, Leukocyte/drug effects , Cytokines/immunology , Dendritic Cells/immunology , Endocytosis/drug effects , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
13.
Eur J Immunol ; 39(9): 2482-91, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19701888

ABSTRACT

Dendritic cells (DC) play a central role in the initiation and regulation of immune responses. Increasing evidence has indicated that manipulation of DC can serve as a therapeutic mechanism for immunomodulation. In this study we tested some unique compounds isolated from Antrodia cinnamomea, a medicinal fungus in Taiwan, on mouse bone marrow-derived DC activation. A triterpenoid methyl antcinate K (me-AntK) promoted DC maturation by enhancing the expression of MHC class II, CD86, and reducing the endocytosis. TNF-alpha, MCP-1, and MIP-1beta were secreted by DC after me-AntK treatment, indicating augmentation of innate immunity by me-AntK. Interestingly, the me-AntK-activated DC induced Ag-specific T-cell proliferation and facilitated Th2 differentiation. Examining signaling responses, we found that me-AntK treatment uniquely activated JNK and ERK in DC. Our results demonstrate that me-AntK is the first natural triterpenoid to promote the ability of DC to prime Th2 responses. This suggests that me-AntK can potentially be applied to enhance immune responses and modulate DC function in immunotherapy.


Subject(s)
Antrodia/chemistry , Dendritic Cells/drug effects , Immunologic Factors/pharmacology , Th2 Cells/drug effects , Triterpenes/pharmacology , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cytokines/biosynthesis , Dendritic Cells/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Immunoglobulin G/blood , Immunologic Factors/isolation & purification , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , MAP Kinase Kinase 4/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/immunology , Th2 Cells/immunology , Triterpenes/isolation & purification
14.
Eur J Immunol ; 38(1): 166-73, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18081038

ABSTRACT

The inhibitory effect of DAP12 on macrophages has been revealed by examining myeloid cells from DAP12-deficient mice. In this report, we demonstrate that both DAP12 and the FcepsilonRIgamma-chain (FcRgamma) are required for negative regulation of TLR responses in bone marrow-derived dendritic cells (DC). Loss of both DAP12 and FcRgamma enhanced the pro-inflammatory cytokine production and maturation of DC after TLR stimulation, resulting in a greater percentage of DC that produced IL-12 p40, TNF, and IL-6, and expressed high levels of MHC class II, CD80, and CD86. Whereas DC lacking only DAP12 showed some increased TLR responses, those lacking only FcRgamma had a greater enhancement of maturation and cytokine production, though to a lesser extent than DC lacking both DAP12 and FcRgamma. Additionally, antigen-specific T cell proliferation was enhanced by DAP12(-/-)FcRgamma(-/-) DC relative to wild-type DC after maturation. Similar to DAP12(-/-)FcRgamma(-/-) DC, Syk-deficient DC also had increased inflammatory cytokine production, maturation, and antigen presentation. These results confirm the inhibitory effect of immunoreceptor tyrosine-based activation motif (ITAM) signaling in myeloid cells and show that DC and macrophages differ in their dependence on the ITAM-containing adapters DAP12 and FcRgamma for negative regulation of TLR signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Antigen Presentation/immunology , Dendritic Cells/immunology , Receptors, IgG/deficiency , Toll-Like Receptors/immunology , Animals , Cytokines/biosynthesis , Dendritic Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Toll-Like Receptors/metabolism
15.
J Med Microbiol ; 50(3): 277-283, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11232775

ABSTRACT

A total of 87 invasive isolates of Haemophilus influenzae isolated throughout Taiwan from 1994 to 1998 was collected; 57 were from children <14 years old. In all, 60.9% of isolates were resistant to ampicillin and produced beta-lactamase. Ribotyping revealed six different profiles in 55 isolates of type b, nine profiles in 10 isolates of non-type b and 12 profiles in 22 isolates of non-typable H. influenzae. Among isolates from 35 cases of meningitis, 30 (86%) were in ribogroups 1, 2 and 3 with >90% genetic similarity. Compared with all the other ribogroups, ribogroups 1, 2 and 3, which encompassed all H. influenzae type b, were significantly more prevalent as a cause of meningitis in children <14 years old. Further subtyping of the predominant ribogroup by pulsed-field gel electrophoresis (PFGE) identified differences of 0-6 bands among these isolates of ribogroup 1, which indicated distant relatedness. Automated ribotyping was found to be a useful method and was less time-consuming for molecular epidemiology studies of H. influenzae. PFGE is suggested as an addition to ribotyping to improve discrimination if H. influenzae type b is involved. Differentiating ribogroups between type b and non-type b H. influenzae by genotyping may help to understand the molecular characteristics of outbreaks, endemicity and value of vaccination. According to the results of ribotyping and PFGE, it seems possible that spread of invasive H. influenzae type b had occurred and ribotyping confirmed that there was no clonal spread of non-type b H. influenzae in Taiwan.


Subject(s)
Electrophoresis, Gel, Pulsed-Field , Haemophilus influenzae/classification , Ribotyping , Adult , Child , Humans , Meningitis, Haemophilus/epidemiology , Taiwan/epidemiology , Time Factors
16.
Microbiology (Reading) ; 144 ( Pt 5): 1257-1262, 1998 May.
Article in English | MEDLINE | ID: mdl-9611800

ABSTRACT

A gene encoding the major antigenic protein of phytoplasma associated with sweet potato witches' broom (SPWB) was cloned and analysed by screening the genomic library of SPWB phytoplasma with monoclonal antibodies for SPWB phytoplasma. The entire predicted structural gene encoded an antigenic protein composed of 172 amino acids with a computed molecular mass of 19.15 kDa and a pl value of 9.78. The -10 region of the promoter and the terminator region of the gene were identified and found to be similar to those of prokaryotes. The hydropathy profile of the deduced amino acid sequence consisted of two distinct regions, a strongly hydrophobic N-terminus and a highly hydrophilic C-terminus. This major antigenic protein was also present in phytoplasma associated with peanut witches' broom (PNWB) and the two showed homology based on the results of Western blot analysis, Southern hybridization, Northern hybridization, primer extension analysis and PCR. The homologous genes of the antigenic protein of SPWB phytoplasma and PNWB phytoplasma were not found in other phytoplasmas tested.


Subject(s)
Antigens, Bacterial/genetics , Genes, Bacterial , Mycoplasma/genetics , Plant Diseases/microbiology , Solanaceae/microbiology , Antibodies, Monoclonal/immunology , Antigens, Bacterial/immunology , Arachis/microbiology , Blotting, Northern , Blotting, Southern , Blotting, Western , Cloning, Molecular , Molecular Sequence Data , Mycoplasma/immunology , Polymerase Chain Reaction , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...