Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Front Immunol ; 13: 838719, 2022.
Article in English | MEDLINE | ID: mdl-35154164

ABSTRACT

The underlying mechanisms of thymocyte development and lineage determination remain incompletely understood, and the emerging evidences demonstrated that RNA binding proteins (RBPs) are deeply involved in governing T cell fate in thymus. Serine/arginine-rich splicing factor 1 (SRSF1), as a classical splicing factor, is a pivotal RBP for gene expression in various biological processes. Our recent study demonstrated that SRSF1 plays essential roles in the development of late thymocytes by modulating the T cell regulatory gene networks post-transcriptionally, which are critical in response to type I interferon signaling for supporting thymocyte maturation. Here, we report SRSF1 also contributes to the determination of the CD8+ T cell fate. By specific ablation of SRSF1 in CD4+CD8+ double positive (DP) thymocytes, we found that SRSF1 deficiency impaired the maturation of late thymocytes and diminished the output of both CD4+ and CD8+ single positive T cells. Interestingly, the ratio of mature CD4+ to CD8+ cells was notably altered and more severe defects were exhibited in CD8+ lineage than those in CD4+ lineage, reflecting the specific function of SRSF1 in CD8+ T cell fate decision. Mechanistically, SRSF1-deficient cells downregulate their expression of Runx3, which is a crucial transcriptional regulator in sustaining CD8+ single positive (SP) thymocyte development and lineage choice. Moreover, forced expression of Runx3 partially rectified the defects in SRSF1-deficient CD8+ thymocyte maturation. Thus, our data uncovered the previous unknown role of SRSF1 in establishment of CD8+ cell identity.


Subject(s)
CD4 Antigens/genetics , CD8-Positive T-Lymphocytes/metabolism , Core Binding Factor Alpha 3 Subunit/metabolism , Serine-Arginine Splicing Factors/deficiency , Thymocytes/metabolism , Animals , CD4 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Core Binding Factor Alpha 3 Subunit/genetics , Down-Regulation , Gene Expression Regulation/immunology , Hematopoiesis , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Serine-Arginine Splicing Factors/genetics
2.
Front Immunol ; 12: 791220, 2021.
Article in English | MEDLINE | ID: mdl-34917097

ABSTRACT

T cell factor 1 (Tcf1) is known as a critical mediator for natural killer (NK) cell development and terminal maturation. However, its essential targets and precise mechanisms involved in early NK progenitors (NKP) are not well clarified. To investigate the role of Tcf1 in NK cells at distinct developmental phases, we employed three kinds of genetic mouse models, namely, Tcf7fl/flVavCre/+, Tcf7fl/flCD122Cre/+ and Tcf7fl/flNcr1Cre/+ mice, respectively. Similar to Tcf1 germline knockout mice, we found notably diminished cell number and defective development in BM NK cells from all strains. In contrast, Tcf7fl/flNcr1Cre/+ mice exhibited modest defects in splenic NK cells compared with those in the other two strains. By analyzing the published ATAC-seq and ChIP-seq data, we found that Tcf1 directly targeted 110 NK cell-related genes which displayed differential accessibility in the absence of Tcf1. Along with this clue, we further confirmed that a series of essential regulators were expressed aberrantly in distinct BM NK subsets with conditional ablating Tcf1 at NKP stage. Eomes, Ets1, Gata3, Ikzf1, Ikzf2, Nfil3, Runx3, Sh2d1a, Slamf6, Tbx21, Tox, and Zeb2 were downregulated, whereas Spi1 and Gzmb were upregulated in distinct NK subsets due to Tcf1 deficiency. The dysregulation of these genes jointly caused severe defects in NK cells lacking Tcf1. Thus, our study identified essential targets of Tcf1 in NK cells, providing new insights into Tcf1-dependent regulatory programs in step-wise governing NK cell development.


Subject(s)
Hepatocyte Nuclear Factor 1-alpha/metabolism , Killer Cells, Natural/physiology , Lymphocyte Subsets/physiology , Lymphoid Progenitor Cells/physiology , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Cell Differentiation , Cells, Cultured , Gene Expression Regulation , Granzymes/genetics , Granzymes/metabolism , Hepatocyte Nuclear Factor 1-alpha/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 1/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
3.
Mol Hum Reprod ; 25(12): 825-837, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31633177

ABSTRACT

Prostaglandin E2 (PGE2) is a hormone with many physiological functions. During pregnancy, it is generally believed that there is a high level of PGE2 at the final stage of pregnancy, which induces the contraction of uterine smooth muscle and promotes the occurrence of childbirth. However, we find that high PGE2 levels are present throughout late pregnancy in mice, not just during childbirth, and that PGE2 deficiency induced by indomethacin during late pregnancy causes damage to the placental labyrinth and eventually leads to abortion. Interestingly, the damage is closely related to inflammation, which involves the role of inflammatory factors produced by the periaortic lymph nodes (PLNs) near the uterus. Further, through RNA sequencing, we reveal that PLNs produce a large amount of interleukin-1ß (IL-1ß) when exposed to PGE2 deficiency, which causes damage to the placental labyrinth, probably via destroying the extracellular matrix. Finally, events leading to abortion following indomethacin administration are effectively prevented by supplementing PGE2 or by PLN removal. These results suggest that high levels of PGE2 during late pregnancy protect fetuses from inflammatory damage related to IL-1ß. This work suggests a new role of PGE2 during late pregnancy and may provide potential therapeutic strategies for pathological pregnancy.


Subject(s)
Abortion, Spontaneous/blood , Chorionic Villi/pathology , Dinoprostone/deficiency , Dinoprostone/metabolism , Interleukin-1beta/metabolism , Animals , C-Reactive Protein , Extracellular Matrix/pathology , Female , Humans , Indomethacin/adverse effects , Inflammation/pathology , Interleukin-1beta/blood , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nerve Tissue Proteins/blood , Pregnancy
4.
FASEB J ; 33(6): 6904-6918, 2019 06.
Article in English | MEDLINE | ID: mdl-30811956

ABSTRACT

Spaceflight leads to health risks including bone demineralization, skeletal muscle atrophy, cardiovascular dysfunction, and disorders of almost all physiologic systems. However, the impacts of microgravity on blood lineage cells and hematopoietic stem cells (HSCs) in vivo are largely unknown. In this study, we analyzed peripheral blood samples from 6 astronauts who had participated in spaceflight missions and found significant changes in several cell populations at different time points. These dynamic alterations of lineage cells and the role of HSCs were further studied in a mouse model, using hindlimb unloading (HU) to simulate microgravity. Large reductions in the frequency of NK cells, B cells, and erythrocyte precursors in the bone marrow of the HU mice were observed, together with an increased frequency of T cells, neutrophils, and HSCs. T cell levels recovered faster than those of B cells and erythrocyte precursors, whereas the recovery rates of NK cells and granulocytes were slow. In addition, competitive reconstitution experiments demonstrated the impaired function of HSCs, although these changes were reversible. Deep sequencing showed changes in the expression of regulatory molecules important for the differentiation of HSCs. This study provides the first determination of altered HSC function under simulated microgravity in vivo. The impairment of HSC function and differentiation provides an explanation for the immune disorders that occur under simulated microgravity. Thus, our findings demonstrated that spaceflight and simulated microgravity disrupt the homeostasis of immune system and cause dynamic alterations on both HSCs and lineage cells.-Cao, D., Song, J., Ling, S., Niu, S., Lu, L., Cui, Z., Li, Y., Hao, S., Zhong, G., Qi, Z., Sun, W., Yuan, X., Li, H., Zhao, D., Jin, X., Liu, C., Wu, X., Kan, G., Cao, H., Kang, Y., Yu, S., Li, Y. Hematopoietic stem cells and lineage cells undergo dynamic alterations under microgravity and recovery conditions.


Subject(s)
Cell Differentiation , Cell Lineage , Hematopoietic Stem Cells/cytology , Hindlimb Suspension/physiology , Homeostasis , Recovery of Function , Weightlessness Simulation , Animals , Astronauts , Erythrocytes/cytology , Humans , Lymphocytes/cytology , Male , Mice , Mice, Inbred C57BL , Neutrophils/cytology , Space Flight
5.
FASEB J ; 33(4): 5615-5625, 2019 04.
Article in English | MEDLINE | ID: mdl-30668923

ABSTRACT

Hematopoietic stem cells (HSCs) have the capacity for self-renewal to maintain the HSCs' pool and the ability for multilineage differentiation, which are responsible for sustained production of multiple blood lineages. The regulation of HSC development is controlled precisely by complex signal networks and hematopoietic microenvironment, which has been termed the HSCs' niche. The Wnt signaling pathway is one of a variety of signaling pathways that have been involved in HSC self-renewal and maintenance. Previous studies are indeterminant on the regulation of adult HSCs upon canonical Wnt signaling pathways because of the different experimental systems and models used. In this study, we generated the conditional knockout Wnt coreceptor low-density lipoprotein receptor-related protein 5 (Lrp5) and low-density lipoprotein receptor-related protein 6 (Lrp6) mice in adult hematopoiesis via Vav-Cre Loxp system. Inactivation of Lrp5 and -6 in a hematopoietic system diminished the pool of HSCs, but there were no obvious defects in mature immune cells. Lrp5 and -6 double deficiency HSCs showed intrinsic defects in self-renewal and differentiation due to reduced proliferation and increased quiescence of the cell cycle. Analysis of HSC gene expression suggested that the quiescence regulators were significantly up-regulated, such as Egr1, Cdkn1a, Nr4a1, Gata2, Junb and Btg2, and the positive cell cycle regulators were correspondingly down-regulated, such as Ccna2 and Ranbp1. Taken together, we investigated the roles of Lrp5 and -6 in HSCs by functional and bioinformatic assays, and we demonstrated that Lrp5 and -6 are required for the self-renewal and differentiation of adult HSCs. The canonical Wnt pathway may contribute to maintaining the HSC pool and regulate the differentiation of adult HSCs by controlling cell cycle gene regulatory module.-Liu, J., Cui, Z., Wang, F., Yao, Y., Yu, G., Liu, J., Cao, D., Niu, S., You, M., Sun, Z., Lian, D., Zhao, T., Kang, Y., Zhao, Y., Xue, H.-H., Yu, S. Lrp5 and Lrp6 are required for maintaining self-renewal and differentiation of hematopoietic stem cells.


Subject(s)
Cell Differentiation/physiology , Hematopoietic Stem Cells/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Animals , Cell Cycle/physiology , Down-Regulation/physiology , Hematopoiesis/physiology , Mice , Stem Cell Niche/physiology , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology
6.
RNA Biol ; 15(12): 1477-1486, 2018.
Article in English | MEDLINE | ID: mdl-30474472

ABSTRACT

Long noncoding RNAs (lncRNAs) are emerging as critical mediators of various biological processes in the immune system. The current data showed that the lncRNA Malat1 is highly expressed in T cell subsets, but the function of Malat1 in T cell remains unclear. In this study, we detected the T cell development and both CD8+ and CD4+ T cell response to LCMV infection using Malat1-/- mice model. To our surprise, there were no significant defects in thymocytes at different developmental stages and the peripheral T cell pool with ablation of Malat1. During LCMV infection, Malat1-/- mice exhibited normal effector and memory CD8+ T cells as well as TFH cells differentiation. Our results indicated that Malat1 is not essential for T cell development and T cell-mediated antiviral response though it expresses at very high level in different T cell populations.


Subject(s)
Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , RNA, Long Noncoding/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Biomarkers , Cell Differentiation , Humans , Immunophenotyping , Mice , Mice, Knockout , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
7.
Sci Rep ; 7(1): 5814, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28724955

ABSTRACT

Currently, chronic hepatitis B virus (HBV) infection remains a serious public health problem in the world. Recombinant HBV vaccine, as a preventive strategy against HBV infection, generates high antibody level, but it is not effective to activate innate and cellular immunity for chronic HBV infection therapy. Lectins from mushroom are natural and active proteins which have been shown important biological functions. However, little is known about the immunological mechanism engaged by mushroom lectins. Here we report that, lectin from Pleurotus ostreatus (POL) stimulated innate response by activating Toll-like receptor 6 signal pathway of dendritic cells. Subsequently POL enhanced HBV specific antibody level and follicular helper T cells response which overcame HBV tolerance in transgenic mice. This study suggests a novel mechanism for POL acting on immune response and a therapeutic approach to break HBV tolerance.


Subject(s)
Agaricales/chemistry , Hepatitis B virus/immunology , Immune Tolerance/drug effects , Lectins/pharmacology , Signal Transduction , Toll-Like Receptor 6/metabolism , Animals , Cell Differentiation/drug effects , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Hepatitis B virus/drug effects , Immunity, Cellular/drug effects , Immunity, Innate/drug effects , Immunization , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes, Helper-Inducer/drug effects
9.
PLoS One ; 11(7): e0160325, 2016.
Article in English | MEDLINE | ID: mdl-27459307

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0154238.].

10.
PLoS One ; 11(5): e0154238, 2016.
Article in English | MEDLINE | ID: mdl-27182980

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most devastating pathogens in the swine industry worldwide. Due to the lack of robust cell lines and small animal models, the pathogenesis of PRRSV infection and mechanism for protective vaccination are still not yet well understood. To obtain useful cell lines, several groups have attempted to construct different transgenic cell lines with three PRRSV receptors: CD163, CD169, and CD151. The results showed that CD163 is essential for PRRSV entry into target cells and replication, and both CD169 and CD151 play key roles during PRRSV infection. However, their interplay and combined effect remains unclear. In this study, we generated transgenic BHK-21 derived cell lines co-expressing different combinations of the three receptors, which were transfected with CD163 alone, or the combination of CD163 and CD169, or the combination of CD163 and CD151, or the combination of CD163, CD169, and CD151 using the PiggyBac transposon system. Our results showed that the synergistic interaction among the three receptors was important to improve the susceptibility of cells during PRRSV infection. Through a series of comparable analyses, we confirmed that the cell line co-expressing triple receptors sustained viral infection and replication, and was superior to the current cell platform used for the PRRSV study, MARC-145 cells. Moreover, we found that PRRSV infection of the transgenic cell lines could trigger IFN-stimulated gene responses similar to those of porcine alveolar macrophages and MARC-145 cells. In summary, we developed a stable transgenic cell line susceptible to PRRSV, which may not only provide a useful tool for virus propagation, vaccine development, and pathogenesis studies, but also establish the foundation for small animal model development.

11.
Biomed Res Int ; 2013: 798164, 2013.
Article in English | MEDLINE | ID: mdl-24308005

ABSTRACT

Protein vaccines combined with adjuvants have been widely used to induce immune responses, especially the humoral immune response, against molecular targets including parasites. Follicular T helper (Tfh) cells are the specialized providers of B-cell help, however, the induction of Tfh cells in protein vaccination has been rarely studied. Here, we report that the Schistosoma japonicum recombinant protein (SjGST-32) combined with tacrolimus (FK506) augmented the induction of Tfh cells, which expressed the canonical markers CXCR5, BCL6, and IL-21, and enhanced the humoral immune responses in BALB/c mice. Furthermore, the expression of IL-21R on germinal center (GC) B cells and memory B cells increased in immunized mice, which indicated that IL-21 from the induced Tfh cells interacted with IL-21R for activation of B cells and maintenance of long-lived humoral immunity. Our results suggest that helminth protein vaccine combined with FK506 induces Tfh cell for stimulating humoral immune responses and inducing long-lived humoral immunity.


Subject(s)
Helminth Proteins/immunology , Schistosoma japonicum/immunology , Schistosomiasis mansoni/immunology , T-Lymphocytes, Helper-Inducer/immunology , Vaccines/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Female , Germinal Center/immunology , Germinal Center/pathology , Helminth Proteins/pharmacology , Immunity, Humoral/drug effects , Immunologic Memory/drug effects , Immunosuppressive Agents/pharmacology , Interleukin-21 Receptor alpha Subunit/immunology , Interleukins/immunology , Mice , Mice, Inbred BALB C , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Schistosoma japonicum/genetics , Schistosomiasis mansoni/genetics , Schistosomiasis mansoni/prevention & control , T-Lymphocytes, Helper-Inducer/pathology , Tacrolimus/pharmacology , Vaccines/pharmacology
12.
PLoS One ; 8(7): e70056, 2013.
Article in English | MEDLINE | ID: mdl-23894591

ABSTRACT

BACKGROUND: Vaccination could induce immune tolerance and protected NOD mice from the development of type I diabetes (T1D). We previously demonstrated that insulin peptide (B9-23) combined with dexamethasone (DEX) stimulated the expansion of antigen specific regulatory T (Treg) cells which in turn effectively prevented T1D in NOD mice. Here, we aimed to investigate the therapeutic effect of tolerogenic vaccination for T1D treatment. METHODOLOGY/PRINCIPAL FINDINGS: The diabetic NOD mice (Blood glucose level ≧250 mg/dl) were treated with B9-23 and DEX twice. The tolerance was restored by blocking maturation of dendritic cells (DCs) and inducing Treg cells in treated NOD mice. Remarkably, the reduction of autoreactive effector memory CD4 T (Tm) cells and the induction of functional effector memory Treg (mTreg) cells contributed to the improvement of T1D in treated NOD mice. CONCLUSIONS/SIGNIFICANCE: Tolerogenic vaccination restored tolerance and ameliorated T1D by suppressing effector CD4 Tm cells and inducing effector mTreg cells. Our findings implicate the potential of tolerogenic vaccination for T1D treatment.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Immune Tolerance , Immunologic Memory , T-Lymphocytes, Regulatory/immunology , Vaccination , Animals , Autoimmunity/drug effects , Cell Differentiation/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dexamethasone/administration & dosage , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/prevention & control , Disease Models, Animal , Female , Insulin/administration & dosage , Insulin/immunology , Mice , Mice, Inbred NOD , Pancreas/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology
13.
Vaccine ; 31(18): 2273-80, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23499522

ABSTRACT

DNA vaccination is a promising strategy for activating immune responses against hepatitis B virus (HBV) infection. However, the accumulated data have shown that DNA vaccination alone generates weak immune responses. To enhance the immunogenicity of HBV DNA vaccine, lectin purified from pleurotus ostreatus (POL) was used as adjuvant of HBV DNA vaccine for C57BL/6 and HBV surface antigen transgenic (HBVsAg-Tg) mice. Our data demonstrate that low dose of POL (1 µg/mouse) in conjunction with HBV DNA vaccine stimulated stronger HBV-specific delayed-type hypersensitivity (DTH) responses and higher HBV-specific IgG level than that in high dose of POL groups (5 µg/mouse and 10 µg/mouse). POL activated strong Th2 and Tc1 cell responses in immunized C57BL/6 and HBVsAg-Tg mice. POL as adjuvant of HBV DNA vaccine effectively enhanced HBV surface protein antibody (HBVsAb) and decreased HBVsAg level for HBV Tg mice treatment. Furthermore, POL infiltrated more lymphocytes excluding Th1, Th2 and Tc1 cell subtypes to liver of HBVsAg-Tg mice. Together, these results suggest that POL as adjuvant enhanced immunogenicity of HBV DNA vaccination and effectively stimulated immune reaponse for HBsAg-Tg mice treatment. Our findings implicate the potential of mushroom lectin as adjuvant of HBV DNA vaccine.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Hepatitis B Vaccines/immunology , Lectins/immunology , Vaccines, DNA/immunology , Adjuvants, Immunologic/pharmacology , Animals , Cell Proliferation , Cytokines/immunology , Dose-Response Relationship, Immunologic , Female , Hepatitis B/immunology , Hepatitis B Antibodies/blood , Hepatitis B Surface Antigens/blood , Hepatitis B Vaccines/genetics , Hypersensitivity, Delayed/immunology , Immunity, Cellular , Lectins/administration & dosage , Liver/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pleurotus/chemistry , T-Lymphocytes, Cytotoxic/immunology , Th2 Cells/immunology
14.
Eur J Immunol ; 43(1): 219-27, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23001956

ABSTRACT

We previously showed that antigen immunization in the presence of the immunosuppressant dexamethasone (a strategy we termed "suppressed immunization") could tolerize established recall responses of T cells. However, the mechanism by which dexamethasone acts as a tolerogenic adjuvant has remained unclear. In the present study, we show that dexamethasone enriches CD11c(lo) CD40(lo) macrophages in a dose-dependent manner in the spleen and peripheral lymph nodes of mice by depleting all other CD11c(+) CD40(+) cells including dendritic cells. The enriched macrophages display a distinct MHC class II (MHC II)(lo) CD86(hi) phenotype. Upon activation by antigen in vivo, CD11c(lo) CD40(lo) macrophages upregulate IL-10, a classic marker for tolerogenic antigen-presenting cells, and elicit a serum IL-10 response. When presenting antigen in vivo, these cells do not elicit recall responses from memory T cells, but rather stimulate the expansion of antigen-specific regulatory T cells. Moreover, the depletion of CD11c(lo) CD40(lo) macrophages during suppressed immunization diminishes the tolerogenic efficacy of the treatment. These results indicate that dexamethasone acts as a tolerogenic adjuvant partly by enriching the CD11c(lo) CD40(lo) tolerogenic macrophages.


Subject(s)
Dexamethasone/administration & dosage , Hypersensitivity, Delayed/drug therapy , Immunosuppressive Agents/administration & dosage , Interleukin-10/immunology , Macrophages/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B7-2 Antigen/metabolism , CD11 Antigens/metabolism , CD40 Antigens/metabolism , Cell Movement/drug effects , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dexamethasone/adverse effects , Histocompatibility Antigens Class II/metabolism , Hypersensitivity, Delayed/immunology , Immune Tolerance , Immunosuppressive Agents/adverse effects , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
15.
PLoS One ; 7(11): e49994, 2012.
Article in English | MEDLINE | ID: mdl-23166807

ABSTRACT

BACKGROUND: Regulatory T (Treg) cells can be induced with DNA vaccinations and protect mice from the development of experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis (MS). Tacrolimus (FK506) has been shown to have functions on inducing immunosuppression and augmenting apoptosis of pathologic T cells in autoimmune disease. Here we examined the therapeutic effect of DNA vaccine in conjunction with FK506 on EAE. METHODOLOGY/PRINCIPAL FINDINGS: After EAE induction, C57BL/6 mice were treated with DNA vaccine in conjunction with FK506. Functional Treg cells were induced in treated EAE mice and suppressed Th1 and Th17 cell responses. Infiltrated CD4 T cells were reduced while Treg cells were induced in spinal cords of treated EAE mice. Remarkably, the activated CD4 T cells augmented apoptosis, but the induced Treg cells resisted apoptosis in treated EAE mice, resulting in alleviation of clinical EAE severity. CONCLUSIONS/SIGNIFICANCE: DNA vaccine in conjunction with FK506 treatment ameliorates EAE by enhancing apoptosis of CD4 T cells and resisting apoptosis of induced Treg cells. Our findings implicate the potential of tolerogenic DNA vaccines for treating MS.


Subject(s)
Apoptosis/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Immune Tolerance/drug effects , Spinal Cord/immunology , T-Lymphocytes, Regulatory/immunology , Tacrolimus/pharmacology , Vaccines, DNA/pharmacology , Analysis of Variance , Animals , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Flow Cytometry , Histological Techniques , Mice , Mice, Inbred C57BL , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , Tacrolimus/immunology , Vaccines, DNA/immunology
16.
PLoS One ; 7(4): e34865, 2012.
Article in English | MEDLINE | ID: mdl-22529945

ABSTRACT

BACKGROUND: H5N1 is a highly pathogenic influenza A virus, which can cause severe illness or even death in humans. Although the widely used killed vaccines are able to provide some protection against infection via neutralizing antibodies, cytotoxic T-lymphocyte responses that are thought to eradicate viral infections are lacking. METHODOLOGY/PRINCIPAL FINDINGS: Aiming to promote cytotoxic responses against H5N1 infection, we extended our previous finding that praziquantel (PZQ) can act as an adjuvant to induce IL-17-producing CD8(+) T cells (Tc17). We found that a single immunization of 57BL/6 mice with killed viral vaccine plus PZQ induced antigen-specific Tc17 cells, some of which also secreted IFN-γ. The induced Tc17 had cytolytic activities. Induction of these cells was impaired in CD8 knockout (KO) or IFN-γ KO mice, and was even lower in IL-17 KO mice. Importantly, the inoculation of killed vaccine with PZQ significantly reduced virus loads in the lung tissues and prolonged survival. Protection against H5N1 virus infection was obtained by adoptively transferring PZQ-primed wild type CD8(+) T cells and this was more effective than transfer of activated IFN-γ KO or IL-17 KO CD8(+) T cells. CONCLUSIONS/SIGNIFICANCE: Our results demonstrated that adding PZQ to killed H5N1 vaccine could promote broad Tc17-mediated cytotoxic T lymphocyte activity, resulting in improved control of highly pathogenic avian influenza virus infection.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Praziquantel/therapeutic use , T-Lymphocytes, Cytotoxic/immunology , Animals , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Female , Humans , Influenza Vaccines/administration & dosage , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/mortality , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
17.
Immunol Invest ; 41(3): 249-60, 2012.
Article in English | MEDLINE | ID: mdl-22221010

ABSTRACT

DNA vaccines have been widely used to induce immune responses against molecular targets. In this study, we explored the possibility of using DNA vaccine combined with the immunosuppressant FK506 (tacrolimus) to antigen-specifically suppress unwanted immune responses and prevent autoimmune ovarian disease. To that end, we immunized C57BL/6 mice with a DNA vaccine encoding mouse zona pellucida 3 (ZP3) together with FK506. The immunization induced ZP3-specific CD4(+)CD25(+)Foxp3(+) regulatory T cells (Treg), which suppressed the induction of ZP3-specific delayed-type hypersensitivity in the animals. Significantly, the immunization also protected the animals from experimentally induced autoimmune ovarian disease. These results suggest that DNA vaccination in the presence of FK506 may be used to induce Treg cells and prevent AOD.


Subject(s)
Autoimmune Diseases/prevention & control , Egg Proteins/metabolism , Hypersensitivity, Delayed/immunology , Membrane Glycoproteins/metabolism , Ovarian Diseases/prevention & control , Receptors, Cell Surface/metabolism , T-Lymphocytes, Regulatory/metabolism , Vaccines, DNA/administration & dosage , Animals , Autoimmune Diseases/immunology , CD4 Antigens/metabolism , Cells, Cultured , Egg Proteins/administration & dosage , Egg Proteins/genetics , Egg Proteins/immunology , Female , Forkhead Transcription Factors/metabolism , Hypersensitivity, Delayed/chemically induced , Hypersensitivity, Delayed/prevention & control , Immune Tolerance , Immunization , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/adverse effects , Interleukin-2 Receptor alpha Subunit/metabolism , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Ovarian Diseases/immunology , Receptors, Cell Surface/administration & dosage , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Tacrolimus/administration & dosage , Tacrolimus/adverse effects , Zona Pellucida Glycoproteins
18.
PLoS One ; 6(10): e25525, 2011.
Article in English | MEDLINE | ID: mdl-21998665

ABSTRACT

BACKGROUND: CD8(+) cytotoxic T lymphocytes (CTLs) are crucial for eliminating hepatitis B virus (HBV) infected cells. DNA vaccination, a novel therapeutic strategy for chronic virus infection, has been shown to induce CTL responses. However, accumulated data have shown that CTLs could not be effectively induced by HBV DNA vaccination. METHODOLOGY/PRINCIPAL FINDINGS: Here, we report that praziquantel (PZQ), an anti-schistoma drug, could act as an adjuvant to overcome the lack of potent CTL responses by HBV DNA vaccination in mice. PZQ in combination with HBV DNA vaccination augmented the induction of CD8(+) T cell-dependent and HBV-specific delayed hypersensitivity responses (DTH) in C57BL/6 mice. Furthermore, the induced CD8(+) T cells consisted of both Tc1 and Tc17 subtypes. By using IFN-γ knockout (KO) mice and IL-17 KO mice, both cytokines were found to be involved in the DTH. The relevance of these findings to HBV immunization was established in HBsAg transgenic mice, in which PZQ also augmented the induction of HBV-specific Tc1 and Tc17 cells and resulted in reduction of HBsAg positive hepatocytes. Adoptive transfer experiments further showed that PZQ-primed CD8(+) T cells from wild type mice, but not the counterpart from IFN-γ KO or IL-17 KO mice, resulted in elimination of HBsAg positive hepatocytes. CONCLUSIONS/SIGNIFICANCE: Our results suggest that PZQ is an effective adjuvant to facilitate Tc1 and Tc17 responses to HBV DNA vaccination, inducing broad CD8(+) T cell-based immunotherapy that breaks tolerance to HBsAg.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Praziquantel/pharmacology , Vaccination , Vaccines, DNA/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Death/drug effects , Cell Death/immunology , Female , Hepatitis B Surface Antigens/genetics , Hepatitis B Surface Antigens/immunology , Hepatitis B Vaccines/immunology , Hepatitis B virus/immunology , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/immunology , Immune Tolerance/drug effects , Immune Tolerance/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic
19.
BMC Immunol ; 12: 27, 2011 May 05.
Article in English | MEDLINE | ID: mdl-21542943

ABSTRACT

BACKGROUND: We previously showed that co-immunization with a protein antigen and a DNA vaccine coding for the same antigen induces CD40 low IL-10 high tolerogenic DCs, which in turn stimulates the expansion of antigen-specific CD4+CD25-Foxp3+ regulatory T cells (CD25- iTreg). However, it was unclear how to choose the antigen sequence to maximize tolerogenic antigen presentation and, consequently, CD25- iTreg induction. RESULTS: In the present study, we demonstrated the requirement of highly antigenic epitopes for CD25- iTreg induction. Firstly, we showed that the induction of CD25- iTreg by tolerogenic DC can be blocked by anti-MHC-II antibody. Next, both the number and the suppressive activity of CD25- iTreg correlated positively with the overt antigenicity of an epitope to activate T cells. Finally, in a mouse model of dermatitis, highly antigenic epitopes derived from a flea allergen not only induced more CD25- iTreg, but also more effectively prevented allergenic reaction to the allergen than did weakly antigenic epitopes. CONCLUSIONS: Our data thus indicate that efficient induction of CD25- iTreg requires highly antigenic peptide epitopes. This finding suggests that highly antigenic epitopes should be used for efficient induction of CD25- iTreg for clinical applications such as flea allergic dermatitis.


Subject(s)
Dendritic Cells/metabolism , Dermatitis, Contact/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , Vaccines, DNA , Adoptive Transfer , Allergens/immunology , Allergens/metabolism , Animals , Antibodies, Blocking/pharmacology , Antigens, CD/metabolism , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/pathology , Dermatitis, Contact/therapy , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Insect Proteins/immunology , Insect Proteins/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Salivary Proteins and Peptides/immunology , Salivary Proteins and Peptides/metabolism , Siphonaptera/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
20.
Vaccine ; 29(29-30): 4862-8, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21481324

ABSTRACT

Cimetidine (CIM) is a histamine H2 receptor inverse agonist used primarily as an anti-stomach acids drug, but recent studies showed that it may also modulate immune responses. To evaluate its potential usefulness as an adjuvant, we determined its immune modulating effects on subunit immunization using an HBV-derived recombinant protein antigen rHBsAg. CIM activated the PI3K-Akt signaling pathway in DCs. As an adjuvant, it activated immunogenic DCs, deactivated tolerogenic T cells (nTreg), and augmented both Th1- and Th2-polarlized immune responses to rHBsAg. As a result, it enhanced both antibody- and cytotoxic T cell-mediated immune responses. Importantly, in comparison with the FDA-approved human adjuvant alum, CIM is superior in its ability to block IL-10 up-regulation and potentiate Th1/Th2 dual polarization. These results suggest that CIM may be a better adjuvant for therapeutic vaccines against chronic viral infection, such as the HBV infection, where dual polarization should allow more effective elimination of infected host cells.


Subject(s)
Cimetidine/administration & dosage , Hepatitis B Surface Antigens/immunology , Immunologic Factors/administration & dosage , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Hepatitis B Antibodies/blood , Mice , Mice, Inbred C57BL , Recombinant Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...