Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters











Publication year range
2.
PLoS One ; 11(10): e0164502, 2016.
Article in English | MEDLINE | ID: mdl-27701472

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0150078.].

3.
PLoS One ; 11(2): e0150078, 2016.
Article in English | MEDLINE | ID: mdl-26919582

ABSTRACT

γδ T cells can either enhance or inhibit an adaptive immune response, but the mechanisms involved are not fully understood. Given that CD73 is the main enzyme responsible for conversion of AMP into the immunosuppressive molecule adenosine, we investigated its role in the regulatory function of γδ T cells in experimental autoimmune uveitis (EAU). We found that γδ T cells expressed different amounts of CD73 during the different stages of EAU and that low CD73 expression on γδ T cells correlated with enhanced Th17 response-promoting activity. Functional comparison of CD73-deficient and wild-type B6 (CD73+/+) mice showed that failure to express CD73 decreased both the enhancing and suppressive effects of γδ T cells on EAU. We also demonstrated that γδ T cells expressed different amounts of CD73 when activated by different pathways, which enabled them to either enhance or inhibit an adaptive immune response. Our results demonstrate that targeting CD73 expression on γδ T cells may allow us to manipulate their pro- or anti-inflammatory effect on Th17 responses.


Subject(s)
5'-Nucleotidase/physiology , Nervous System Autoimmune Disease, Experimental/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Uveitis/immunology , 5'-Nucleotidase/biosynthesis , 5'-Nucleotidase/deficiency , 5'-Nucleotidase/genetics , Adenosine/metabolism , Adenosine Monophosphate/metabolism , Animals , Cells, Cultured , Dendritic Cells/immunology , Eye Proteins/immunology , Eye Proteins/toxicity , Female , Gene Expression Regulation/immunology , Interferon-gamma/blood , Interferon-gamma/deficiency , Interleukin-17/blood , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Nervous System Autoimmune Disease, Experimental/enzymology , Peptide Fragments/immunology , Peptide Fragments/toxicity , Receptors, Antigen, T-Cell, gamma-delta/analysis , Receptors, Antigen, T-Cell, gamma-delta/deficiency , Retinol-Binding Proteins/immunology , Retinol-Binding Proteins/toxicity , T-Lymphocyte Subsets/enzymology , T-Lymphocytes, Regulatory/enzymology , Th1 Cells/immunology , Th17 Cells/immunology , Uveitis/enzymology
4.
J Immunol ; 196(6): 2646-54, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26856700

ABSTRACT

Adenosine is an important regulator of the immune response, and adenosine deaminase (ADA) inhibits this regulatory effect by converting adenosine into functionally inactive molecules. Studies showed that adenosine receptor agonists can be anti- or proinflammatory. Clarification of the mechanisms that cause these opposing effects should provide a better guide for therapeutic intervention. In this study, we investigated the effect of ADA on the development of experimental autoimmune uveitis (EAU) induced by immunizing EAU-prone mice with a known uveitogenic peptide, IRBP1-20. Our results showed that the effective time to administer a single dose of ADA to suppress induction of EAU was 8-14 d postimmunization, shortly before EAU expression; however, ADA treatment at other time points exacerbated disease. ADA preferentially inhibited Th17 responses, and this effect was γδ T cell dependent. Our results demonstrated that the existing immune status strongly influences the anti- or proinflammatory effects of ADA. Our observations should help to improve the design of ADA- and adenosine receptor-targeted therapies.


Subject(s)
Adenosine Deaminase/administration & dosage , Autoimmune Diseases/drug therapy , Immunologic Factors/administration & dosage , Th17 Cells/drug effects , Uveitis/drug therapy , Animals , Autoimmune Diseases/immunology , Cells, Cultured , Eye Proteins/immunology , Female , Humans , Immunization , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Molecular Targeted Therapy , Peptide Fragments/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Retinol-Binding Proteins/immunology , Th17 Cells/immunology , Uveitis/immunology
5.
PLoS One ; 10(7): e0132348, 2015.
Article in English | MEDLINE | ID: mdl-26147733

ABSTRACT

We have recently reported that, although adenosine receptor (AR) agonists have a suppressive effect on Th1 autoreactive T cells, their effect on Th17 autoreactive T cells and γδ T cells is stimulatory and this effect is mainly mediated via A2A adenosine receptors (A2ARs). In this study, we further demonstrate that treatment of C57BL/6 (B6) mice with a selective A2B adenosine receptor (A2BR) agonist greatly enhanced the development of experimental autoimmune uveitis (EAU), whereas treatment with an A2BR antagonist significantly ameliorated severity of EAU. The A2BR agonist-treated mice showed augmented Th17, but not Th1, responses. Mechanistic studies showed that the A2BR agonist-induced enhancement of the Th17 response was significantly lower when TCR-δ-/- mice received the same treatment and that transfer of γδ T cells into TCR-δ-/- mice partially restored this effect. We also showed that dendritic cells (DCs) from A2BR agonist-treated mice showed a significantly increased ability to activate γδ T cells and Th17 autoreactive T cells. Thus, our previous studies have shown that, in EAU, activated γδ T cells possess greatly increased ability to enhance Th17 autoimmune responses. In the present study, we showed that exposure of DCs to A2BR agonist facilitated γδ T cell activation, leading to augmented Th17 responses and progressive EAU development. Our results further support our previous finding that AR agonists have distinct effects on Th1 and Th17 autoimmune responses.


Subject(s)
Adenosine A2 Receptor Agonists/pharmacology , Autoimmune Diseases/immunology , Dendritic Cells/immunology , Receptor, Adenosine A2B/immunology , Th17 Cells/immunology , Uveitis/immunology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Dendritic Cells/pathology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Receptor, Adenosine A2B/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/pathology , Uveitis/genetics , Uveitis/pathology
6.
Immun Inflamm Dis ; 3(4): 360-73, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26734458

ABSTRACT

Adenosine is one of the major molecules associated with inflammation. We have previously reported that an adenosine receptor (AR) agonist has an enhancing effect on Th17 autoimmune responses, even though it suppressed Th1 responses. To determine the mechanism involved, we have examined the effect of AR agonists on mouse bone marrow dendritic cell (BMDC) differentiation and function. We show that mouse bone marrow cells (BMCs) differentiated into CD11c(+)Gr-1(+) dentritic cells (DCs) when cultured in granulocyte macrophage colony-stimulating factor (GM-CSF)-containing medium containing an AR agonist. The non-selective AR agonist NECA and an A2BR-specific agonist had a similar effect, and the effect of NECA could be blocked by an A2BR-specific antagonist. Unlike CD11c(+)Gr-1(-) BMDCs, which have a greater stimulatory effect on Th1 T cells than Th17 cells, CD11c(+)Gr-1(+) BMDCs had a greater stimulatory effect on Th17 autoreactive T cells than on Th1 autoreactive T cells and this effect depended on γδ T cell activation.

7.
J Immunol ; 193(11): 5498-505, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25367119

ABSTRACT

Adenosine is a key endogenous signaling molecule that regulates a wide range of physiological functions, including immune system function and inflammation. Studies have shown that adenosine receptor (AR) agonists can be either anti-inflammatory or proinflammatory in immune responses and in inflammation, and the clarification of the mechanisms causing these opposing effects should provide a better guide for therapeutic intervention. Whereas previous studies mostly examined the effects of AR agonists on Th1-type immune responses, in this study, we compared their effect on Th17 and Th1 autoimmune responses in experimental autoimmune uveitis, a mouse model of human uveitis induced by immunization with the human interphotoreceptor retinoid-binding protein peptides 1-20. We showed that injection of mice with a nonselective AR agonist, 5'-N-ethylcarboxamidoadenosine (NECA), at an early stage after immunization had an inhibitory effect on both Th1 and Th17 responses, whereas injection of the same amount of NECA at a late stage inhibited the Th1 response but had an enhancing effect on the Th17 response. We also showed that the effects of NECA on Th1 and Th17 responses were completely dissociated, that the enhancing effect of NECA on Th17 responses was modulated by γδ T cells, and that the response of γδ T cells to NECA was determined by their activation status. We conclude that the inflammatory environment has a strong impact on converting the effect of AR agonist on the Th17 autoimmune response from anti-inflammatory to proinflammatory. Our observation should help in the designing of better AR-targeted therapies.


Subject(s)
Adenosine-5'-(N-ethylcarboxamide)/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Autoimmune Diseases/immunology , Inflammation Mediators/administration & dosage , Purinergic P1 Receptor Agonists/administration & dosage , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Th1 Cells/immunology , Th17 Cells/immunology , Uveitis/immunology , Animals , Autoantigens/immunology , Autoimmune Diseases/chemically induced , Autoimmune Diseases/therapy , Cells, Cultured , Eye Proteins/immunology , Female , Humans , Immunomodulation/drug effects , Immunomodulation/genetics , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Peptide Fragments/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Retinol-Binding Proteins/immunology , Th1 Cells/drug effects , Th17 Cells/drug effects , Uveitis/chemically induced , Uveitis/therapy
8.
PLoS One ; 9(9): e108932, 2014.
Article in English | MEDLINE | ID: mdl-25268760

ABSTRACT

The adenosine A2A receptor (A2AR), the main functional adenosine receptor on murine T cells, plays a unique role in the attenuation of inflammation and tissue damage in vivo. Here, we showed that, of the immune cell types tested, activated γδ T cells expressed the highest levels of A2AR mRNA and that A2AR ligation inhibited αß T cell activation, but enhanced γδ T cell activation. We also showed that the inhibitory effect of an adenosine receptor agonist on autoreactive T cells was prevented by addition of a low percentage of activated γδ T cells. Furthermore, compared to resting cells, activated γδ T cells expressed significantly lower levels of CD73, an enzyme involved in the generation of extracellular adenosine. Exogenous AMP had a significant inhibitory effect on autoreactive T cell responses, but only in the presence of CD73+ γδ T cells, and this effect was abolished by a CD73 inhibitor. Our results show that expression of increased amounts of A2AR allows γδ T cells to bind adenosine and thereby attenuate its suppressive effect, while decreased expression of CD73 results in less generation of adenosine in the inflammatory site. Together, these events allow activated γδ T cells to acquire increased proinflammatory activity, leading to augmented autoimmune responses.


Subject(s)
5'-Nucleotidase/metabolism , Receptor, Adenosine A2A/metabolism , T-Lymphocytes/metabolism , 5'-Nucleotidase/antagonists & inhibitors , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/pharmacology , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine Monophosphate/pharmacology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/pharmacology , Dinucleoside Phosphates/pharmacology , Female , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Phenethylamines/pharmacology , RNA, Messenger/metabolism , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/genetics , Signal Transduction/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/drug effects
9.
Mol Cell Biochem ; 389(1-2): 141-50, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24481752

ABSTRACT

We have previously identified a novel Trß isoform (TrßΔ) in the rat, in which a novel exon N (108 bps) was found between exon 3 and exon 4 of TrßΔ, which represents the only difference between TrßΔ and Trß1. In this study, we searched for an elongated Trß2-like subtype with one additional exon N. We successfully isolated the entire mRNA/cDNA of a novel elongated Trß2 isoform via PCR in the rat pituitary gland. The mRNA/cDNA was only 108 bps (exon N) longer than that Trß2, and the extension of the sequence was between exon 3 and 4 of Trß. The whole sequence of this novel Trß isoform has been published in NCBI GenBank (HM043807.1); it is named TRbeta2Delta (Trß2Δ). In adult rat pituitary tissue, quantitative real-time RT-PCR analysis showed that the mRNA levels of Trß2Δ and Trß2 were roughly equal (P > 0.05). We cloned, expressed, and purified the His-Trß2Δ protein [recombinant TRß2Δ (rTRß2Δ)]. SDS-PAGE and western blotting revealed that the molecular weight of rTRß2Δ was 58.2 kDa. Using a radioligand binding assay and an electrophoretic mobility shift assay, rTRß2Δ-bound T3 with high affinity and recognized thyroid hormone response element (TRE) binding sites. Finally, in vitro transfection experiments further confirmed that rTRß2Δ binding T3 significantly promotes the transcription of target genes via the TRE. Here, we have provided evidence suggesting that rTRß2Δ is a novel functional TR isoform.


Subject(s)
Pituitary Gland/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Receptors beta/metabolism , Animals , Binding Sites/genetics , COS Cells , Chlorocebus aethiops , Cloning, Molecular , DNA, Complementary/genetics , Ligands , RNA, Messenger/genetics , Rats , Transcription, Genetic/genetics , Transfection/methods
10.
Eur J Pharm Biopharm ; 85(3 Pt A): 726-35, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24055691

ABSTRACT

The local interaction between retinal pigment epithelium (RPE) and immigrated effector T cells is crucial for the pathogenesis of autoimmune uveitis. After being activated by the pattern recognition receptors (PRRs) signaling pathway, RPE can present the antigen reactivated invading autoreactive T cells, resulting in uveitis. In the present study, we showed that the transfection of chitosan-loaded TLR3-siRNA toward RPE could effectively remit experimental autoimmune uveitis (EAU) in B10RIII mice. Initially, we verified the constitutive expression of Tlr3 in RPE at high levels, which was not altered in response to TNFα, IFNγ and IL-17A treatments. Compared with other TLRs, the activation of TLR3 signaling following polyIC treatment resulted in increased IL-6 and IFNγ secretion from and MHCII expression on RPE. It is polyIC-, but not other TLR ligands, treated RPE showed significant synergetic effect with IL-17 on stimulating RPE secreting CXCL8 and CCl2, which might be resulted from elevated Il17ra expression in RPE following polyIC treatment. Furthermore, polyIC-treated RPE caused a robust stimulation of differentiation of CD4 cell toward Th1 or Th17 cells, in addition to the secretion of the cytokines IFNγ and IL-17. The in vitro knockdown of TLR3 expression in RPE by chitosan/TLR3-siRNA transfection could effectively block polyIC-induced MHCII expression, pro-inflammatory cytokine secretion and autoreactive CD4 cell activation. Studies conducted in firefly luciferase gene transgenic mice demonstrated that the subretinal CS/Luci-siRNA transfection specifically reduced the luciferase activity in RPE but not in the liver and spleen. Finally, the CS/TLR3-siRNA was locally administered in the EAU induced B10RIII mice. The results revealed that chitosan-mediated TLR3-siRNA transfection had a significant therapeutic effect on either delaying the outbreak or remitting the severity of uveitis.


Subject(s)
Chitosan/chemistry , RNA, Small Interfering/administration & dosage , Toll-Like Receptor 3/genetics , Uveitis/therapy , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Autoimmune Diseases/therapy , CD4 Antigens/metabolism , Cytokines/immunology , Disease Models, Animal , Female , Gene Knockdown Techniques , Interferon-gamma/immunology , Interleukin-6/immunology , Luciferases, Firefly/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Retinal Pigment Epithelium/metabolism , Severity of Illness Index , Signal Transduction , Transfection , Uveitis/immunology , Uveitis/pathology
11.
J Immunol ; 191(3): 1118-25, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23797670

ABSTRACT

We have previously reported that, depending on their activation status, mouse γδ T cells can either enhance or inhibit the activity of IL-17(+) autoreactive T cells in experimental autoimmune uveitis. In this study, we showed that γδ T cells in naive C57BL/6 (B6) mouse do not express the IL-23R, whereas in immunized mice, it is expressed on >50% of γδ T cells. In vitro studies showed that IL-23R expression on γδ T cells is modulated by their state of activation, as weakly activated γδ T cells expressed the IL-23R, but highly activated γδ T cells did not. Functional studies showed that IL-23R(+) γδ T cells had the strongest suppressive effect on IL-17(+) autoreactive T cells, and that this effect was inhibited when the IL-23R was blocked by anti-IL-23R Ab or in the presence of excessive amounts of exogenous IL-23. We conclude that the balance between the enhancing and inhibitory effects of γδ T cells is regulated by their level of IL-23R expression. The expression of variable IL-23R levels allows γδ T cells to have different regulatory effects on adaptive immune responses, conceivably as a result of αß and γδ T cells competing for IL-23.


Subject(s)
Autoimmune Diseases/immunology , Interleukin-17/metabolism , Interleukin-23/pharmacology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Interleukin/immunology , T-Lymphocytes/immunology , Uveitis/immunology , Animals , Antibodies/immunology , Autoimmunity , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Interleukin/biosynthesis , T-Lymphocytes/metabolism , Uveitis/chemically induced
12.
Invest Ophthalmol Vis Sci ; 54(5): 3493-503, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23611991

ABSTRACT

PURPOSE: We determined the mechanism by which all-trans retinoic acid (ATRA) inhibits experimental autoimmune uveitis (EAU) and determined the role of γδ T cells in this autoimmune disease. METHODS: C57BL/6 (B6) mice were immunized with the uveitogenic, interphotoreceptor retinoid-binding protein1-20 peptide (IRBP1-20) in complete Freund's adjuvant (CFA), with or without a preceding ATRA treatment. Responses and pathogenic activity of Th1- and Th17-autoreactive T cells were compared, and the effects of ATRA on γδ T cells and CD25(+) dendritic cell (DC) subset were determined. Interactions among uveitogenic T cells, DC subsets, and γδ T cells were investigated. RESULTS: Administration of ATRA to B6 mice in which EAU was induced suppressed the response of Th17 autoreactive T cells, which was associated with decreased generation of the CD25(+) DC subset and suppressed activation of γδ T cells. Adoptively transferred γδ T cells isolated from ATRA-treated mice showed a diminished ability to promote the activation of Th17 autoreactive T cells in vitro and in vivo compared to γδ T cells from untreated donors. CONCLUSIONS: ATRA inhibits the expansion of CD25(+) DCs and γδ T-cell activation, thereby restraining the Th17 autoreactive T-cell response.


Subject(s)
Autoimmune Diseases/drug therapy , Dendritic Cells/drug effects , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Tretinoin/pharmacology , Uveitis/drug therapy , Animals , Antineoplastic Agents/pharmacology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Interleukin-17/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/metabolism , Uveitis/immunology , Uveitis/pathology
13.
J Immunol ; 188(11): 5785-91, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22539790

ABSTRACT

In the current study, we showed that in vivo administration of an anti-CD25 Ab (PC61) decreased the Th17 response in C57BL/6 mice immunized with the uveitogenic peptide interphotoreceptor retinoid-binding protein, while enhancing the autoreactive Th1 response. The depressed Th17 response was closely associated with decreased numbers of a splenic dendritic cell (DC) subset expressing CD11c(+)CD3(-)CD25(+) and decreased expansion of γδ T cells. We demonstrated that ablation of the CD25(+) DC subset accounted for the decreased activation and the expansion of γδ T cells, leading to decreased activation of IL-17(+) interphotoreceptor retinoid-binding protein-specific T cells. Our results show that an enhanced Th17 response in an autoimmune disease is associated with the appearance of a DC subset expressing CD25 and that treatment of mice with anti-CD25 Ab causes functional alterations in a number of immune cell types, namely DCs and γδ T cells, in addition to CD25(+)αßTCR(+) regulatory T cells.


Subject(s)
Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Cell Differentiation/immunology , Dendritic Cells/immunology , Interleukin-2 Receptor alpha Subunit/biosynthesis , Th17 Cells/immunology , Uveitis/immunology , Uveitis/pathology , Animals , Autoimmune Diseases/metabolism , Dendritic Cells/metabolism , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Female , Interleukin-2 Receptor alpha Subunit/physiology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Th17 Cells/pathology , Uveitis/metabolism
14.
Mol Immunol ; 50(3): 125-33, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22301006

ABSTRACT

We investigated the in vivo priming of IL-17(+) autoreactive T cells in experimental autoimmune uveitis-prone C57BL/6 (B6) and B10RIII mice using a combination of approaches, including limiting dilution assay. High numbers of in vivo primed IL-17(+) interphotoreceptor retinoid-binding protein (IRBP)-specific T cells were found in mice immunized with a uveitogenic peptide emulsified in CFA, but not the same peptide emulsified in IFA. Both in vitro and in vivo, at least part of the effect of mycobacterial antigen in CFA could be replaced by TLR2 or TLR4 ligands. TCR-δ(-/-) mice immunized with IRBP peptide in CFA generated significantly lower numbers of IL-17(+) T cells than immunized wild-type B6 mice. Administration of a small number of activated γδ T cells to TCR-δ(-/-) mice significantly increased the number of IL-17(+), but not IFN-γ(+), IRBP-specific T cells in these mice. γδ T cells from CFA- or IFA plus TLR ligand-treated mice were activated and injection of naïve TCR-δ(-/-) mice with γδ T cells from TLR ligand-treated, but not untreated, B6 mice promoted the in vivo priming of IL-17(+) IRBP-reactive T cells. In conclusion, in vivo priming of IL-17(+) uveitogenic T cells in mice is significantly affected by TLR ligation, and is also influenced by activated γδ T cells.


Subject(s)
Interleukin-17/immunology , Lymphocyte Activation/drug effects , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Toll-Like Receptor 2/agonists , Toll-Like Receptor 4/agonists , Animals , Cell Proliferation , Female , Ligands , Mice , Receptors, Antigen, T-Cell, gamma-delta/deficiency , T-Lymphocytes/cytology , T-Lymphocytes/drug effects
15.
Invest Ophthalmol Vis Sci ; 53(2): 897-905, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22247477

ABSTRACT

PURPOSE: To characterize antigen-specific and bystander IL-17+ T cells induced in immunized mice. METHODS: C57BL/6 (B6) mice were immunized with the uveitogenic peptide IRBP1₋20 in either incomplete (IFA) or complete (CFA) Freund's adjuvant. In vivo-primed T cells were stimulated with syngeneic APCs, with or without the immunizing peptide, under polarizing conditions. Activated T cells were analyzed for expression and production of IL-17. RESULTS: B6 mice immunized with the uveitogenic peptide IRBP1₋20 generated two types of IL-17+ T cell: one specific for the immunizing autoantigen (IRBP-Th17) and a much more abundant type (bystander-Th17) that is not reactive with the immunizing antigen. The bystander-Th17 can be demonstrated when in vivo-primed T cells are cultured in Th17-polarizing conditions in the absence of antigen stimulation. Increased expansion of both types of Th17 cells was seen in mice immunized with IRBP1₋20/CFA, but not with IRBP1₋20/IFA. Both T-cell types produced IL-17, IL-22, and IFN-γ, but only bystander Th17 cells produced IL-10. Addition to culture medium of IL-6 and TGF-ß1 caused more activation of bystander-Th17 T cells than IRBP-Th17 cells. When adoptively transferred into syngeneic naïve mice, the bystander-Th17 cells neutralized the pathogenic activity of the IRBP-Th17 cells. CONCLUSIONS: A procedure commonly used to induce autoimmune disease promotes two functionally antagonistic types of IL-17+ T cells, and the pathogenic type is restricted to the population that specifically responds to the immunizing autoantigen. Molecular components of the CFA, rather than the immunizing peptide, promote the generation of both types of IL-17+ T cells.


Subject(s)
Autoimmune Diseases/immunology , Bystander Effect/immunology , Interleukin-17/immunology , Lymphocyte Activation/immunology , Th17 Cells/immunology , Uveitis/immunology , Animals , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Disease Models, Animal , Female , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Uveitis/pathology
16.
Int J Pharm ; 422(1-2): 445-53, 2012 Jan 17.
Article in English | MEDLINE | ID: mdl-22067703

ABSTRACT

Small interfering RNA (siRNA) has been widely investigated as a potential therapeutic approach for diseases with genetic defects. However, its application was greatly hampered by the rapid degradation and poor cellular uptake. Recently, chitosan (CS) and its derivant have been considered as a promising siRNA transporter with the advantages of low toxicity, good biodegradability and biocompatibility. Chitosan of different molecular weight (Mw) and degrees of deacetylation (DD) showed significantly varied target gene silencing efficacy, and it is still not well clarified how these characteristics influence CS mediated siRNA transfection. To compare the aspects of cell permeability and intracellular unpacking of CS/siRNA complex on the effect of CS/siRNA transfection. A radiolabeled siRNA, targeting firefly luciferase gene, was loaded by chitosan of different molecular weight (varying from 2000 to 800,000 Da) and subjected to the transfection against MDA-MB-231/Luc human breast cancer cell line which stably expressed knocked in firefly Luciferase reporter gene. Following transfection intracellular radioactivity was measured to represent cell entrance ability of the CS/siRNA, while, luciferase activity in the cell lysate was also determined to reflect target gene silencing effect. The results revealed that although low molecular weight chitosan (LMWC) condensed siRNA has the highest cell permeability of almost two folds of medium molecular weight chitosan and lipofactamine, its target gene silencing effect is really low of almost eight times less than lipofectamine. This conspicuous contradiction gave us the hypothesis that LMWC generated more condensed CS/siRNA complex to facilitate cell entrance but the tight electrostatic interaction probably limited intracellular siRNA unpacking as well and unfavorably hindered target gene silencing as the final consequence. To approve this hypothesis a phosphorylatable short peptide conjugated LMWC was adopt to promote intracellular siRNA unpacking. Which was demonstrated of perfect target gene knock down ability to the extent of being even superior to lipofactamine 2000. In a conclusion, low molecular weight chitosan has the great potential to be an ideal siRNA vehicle if the issue of siRNA unpacking could be properly resolved.


Subject(s)
Chitosan/metabolism , Peptides/chemistry , RNA Interference , RNA, Small Interfering/metabolism , Transfection/methods , Acetylation , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Chitosan/chemistry , Female , Humans , Lipids/chemistry , Luciferases/biosynthesis , Luciferases/genetics , Molecular Weight , Nanoparticles , Nucleic Acid Conformation , Particle Size , Permeability , Phosphorylation , RNA, Small Interfering/chemistry
17.
Int J Pharm ; 413(1-2): 254-9, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21536119

ABSTRACT

Previously, we had reported improving transfection efficiency of the chitosan-plasmid DNA (CS/pDNA)complex via enhancing intracellular unpacking of the exogene by the utilization of phosphorylatable short peptide conjugated chitosan (pSP-CS). In this article, we addressed a novel strategy of nucleus localization signal linked nucleic kinase substrate short peptide (NNS) modification for further optimization of the transfection efficiency. NNS, consisting of "PKKRKVREEAIKFSEEQRFRR", contained a SV40 nucleus localization signal and a potentially phosphorylatable serine residue. The short peptide could be selectively phosphorylated in the nucleus in various mammalian cells. This phosphorylatable NNS (pNNS) was conjugated to chitosan and combined with Cy3 fluorescence labeled plasmid DNA to form a pNNS-CS/pDNA complex. In vitro phosphorylation and DNA releasing assays verified that pNNS could be effectively and selectively phosphorylated by nucleic lysate, hence promoting pDNA unpacking from the complex. Thereafter, C2C12 myoblast cells were transfected. Nuclear localization of the pDNA was represented by the fluorescence in the nucleus and transfection efficiency was determined by the expression of the luciferase reporter gene, which is carried by the plasmid DNA. The results revealed that, compared with lipofactamine2000 and the previously reported pSP-CS, pNNS-CS could transport more pDNA into the nucleus and intensively augment luciferase reporter gene expression. In conclusion, nucleus localization and unpacking from the delivery vector are both critical factors in influencing exogene expression, and pNNS modification is valuable in improving transfection efficacy of the chitosan.


Subject(s)
Chitosan/metabolism , Drug Compounding/methods , Intranuclear Space/metabolism , Nuclear Localization Signals/metabolism , Transfection/methods , Chitosan/chemistry , DNA/metabolism , Drug Carriers , Drug Delivery Systems , Genes, Reporter , Genetic Vectors , Indicators and Reagents/metabolism , Lipids/genetics , Lipids/pharmacology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peptides/analysis , Peptides/genetics , Peptides/metabolism , Phosphorylation , Plasmids/genetics , Plasmids/metabolism
18.
Yi Chuan ; 33(3): 246-50, 2011 Mar.
Article in Chinese | MEDLINE | ID: mdl-21402532

ABSTRACT

Retinoid X receptor-α (RXR-α), a member of nuclear receptor family, is capable of mediating retinoid signaling pathways and plays a critical role in regulating target gene transcription. To further study the function of RXR-α, abundant of recombinant RXR-α protein in hand is necessary. In this study an intact RXR-α coding sequence was amplified by RT-PCR and subsequently inserted into expression plasmid vector pQE-30Xa to form the recombinant construct of pQE-30Xa/RXR-α. Thereafter, competent bacteria Escherichia coli M15 [PREP4] was transformed and the expression of RXR-α was induced by adding IPTG to the medium. Bacterially expressed recombinant RXR-α was purified by Ni-NTA affinity chromatography and verified by SDS-PAGE and Western blotting analyses. The results showed that a protein, with the molecular mass around 50 kDa, could be selectively recognized by anti-RXR-α antibody. Co-immunoprecipitation assay indicated that this recombinant RXR-α could effectively bind TRß1 to form a heterodimer, which could specifically bind the target DNA fragment. This was confirmed by EMSA. In conclusion, the recombinant human retinoid X receptor-α was prepared successfully, which makes a basic for further study of its function.


Subject(s)
Retinoid X Receptor alpha/genetics , Retinoid X Receptor alpha/metabolism , Thyroid Hormone Receptors beta/metabolism , Cell Line , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Gene Expression , Humans , Immunoprecipitation , Nitrilotriacetic Acid/metabolism , Plasmids/genetics , Polymerase Chain Reaction , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Restriction Mapping , Retinoid X Receptor alpha/chemistry , Retinoid X Receptor alpha/isolation & purification , Thyroid Hormone Receptors beta/chemistry
19.
Int J Pharm ; 397(1-2): 206-10, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20633618

ABSTRACT

Previously, we had demonstrated that enhancing intracellular unpacking of exogene from its chitosan carrier by promoting chitosan degradation could markedly improve transfection efficiency of the CS/DNA complex. In this article we addressed a novel strategy of phosphorylatable short peptide modification for further facilitating intracellular DNA unpacking and optimizing transfection efficiency of the CS/DNA complex. A short peptide (SP) with the amino acid composition of "LLLRRRDNEY*FY*VRRLL" containing two potentially phosphorylatable tyrosine residues was synthesized. The short peptide could be phosphorylated by constitutively expressed cytoplasmic protein kinase Jak2. The SP was conjugated to chitosan and combined with GFP/luciferase reporter gene plasmid DNA to form SP-CS/DNA complex. In vitro phosphorylation and DNA releasing assays verified that mammalian cell lysate could effectively phosphorylate SP and hence promote plasmid DNA unpacking from the SP-CS carrier. Thereafter, C2C12 myoblast cells were transfected by SP-CS/DNA and the transfection efficiency was presented by the expression of GFP and luciferase reporters. Further more, multiple cell lines were transfected by SP-CS/DNA complexes loading luciferase reporter gene. Results revealed that, compared with CS, SP-CS could intensively augment the transfection efficiency to the level of near lipofectamine 2000.


Subject(s)
Chitosan , DNA , Transfection , Animals , Cell Line , Cell Line, Tumor , Chitosan/chemistry , Chitosan/metabolism , DNA/chemistry , DNA/genetics , DNA/metabolism , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lipids , Luciferases/genetics , Mice , Nanoparticles , Particle Size , Phosphorylation , Plasmids
20.
Biomaterials ; 31(1): 144-55, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19783298

ABSTRACT

In this work, thermoresponsive diblock copolymers, poly[2-(2-methoxyethoxy) ethyl methacrylate]-b-poly(2-hydroxyethyl methacrylate) (PMEO(2)MA-b-PHEMA) with low polydispersity were synthesized by atomic transfer radical polymerization(ATRP). Low molecular weight (LWM) polyethylenimine (PEI, 1200Da) was then grafted to 1,1'-carbonyldiimidazole (CDI)-activated PMEO(2)MA-b-PHEMA to fabricate PEI-g-(PMEO(2)MA-b-PHEMA) (PEIMH) copolymer vectors. The LCSTs of PEIMHs with 3 and 8 grafted PEI side chains, separately termed as PEIMH-1 and PEIMH-2, were 32.5 and 38.7 degrees C in PBS solution. Variable temperature agarose retardation, Zeta potential and time-resolved fluorescence assays were performed to elucidate the temperature sensitive DNA condensation. It showed that DNA was condensed more efficiently by PEIMH, and the collapse of PMEO(2)MA chains led to more exposure of surface positive charges of PEIMH-1/pDNA complexes while temperature was above LCST. Variable temperature time-resolved fluorescence measurement of lifetimes of bound and free ethidium bromide (EB) unveiled that the population of EB at different states was dependent on temperature. At a temperature above LCST, the collapsed PMEO(2)MA polymer chains squeezed the loosely bound EB out of complex to become free species; thereby DNA was more tightly packaged by PEIMH-1. Temporary cooling was shown to improve the transfection efficiency of PEIMH-1 in COS-7 and HEK293 cell lines. The variable temperature protocol is more efficient in improving gene expression level in HEK293 cells. The transfection efficiency was equivalent or superior to that of PEI25K at an optimal weight ratio of vector/DNA. Furthermore, the cytotoxicity of PEIMH-1 was considerably lower than that of control PEI25K.


Subject(s)
DNA/chemistry , Genetic Vectors , Polyethyleneimine/chemistry , Polyhydroxyethyl Methacrylate/chemistry , Transfection , Adult , Cells, Cultured , Culture Media , DNA/genetics , Female , Humans , Male , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL