Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Sci Rep ; 10(1): 869, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31964951

ABSTRACT

Obesity is a major health problem and is known to be closely associated with metabolic diseases. Abnormal hepatic accumulation of fat causes fatty liver or hepatic steatosis, and long-term consumption of a high-fat diet is known to be a key obesity-causing factor. Recent studies have demonstrated that probiotics such as Lactobacillus strains, exert an anti-obesity effect by regulating adipogenesis. However, it is still unknown how the consumption of probiotics can reduce abdominal fat volume by regulating the hepatic expression of lipogenic genes. Therefore, we evaluated the effect of long-term ingestion of L. plantarum LMT1-48 on the expression of lipogenic genes in high-fat diet (HFD)-fed mice. We observed that treatment of 3T3-L1 adipocytes with L. plantarum LMT1-48 extract inhibited their differentiation and lipid accumulation by downregulating lipogenic genes, namely, PPARγ, C/EBPα, FAS, and FABP4. Interestingly, administration of L. plantarum LMT1-48 reduced liver weight and liver triglycerides concurrently with the downregulation of the lipogenic genes PPARγ, HSL, SCD-1, and FAT/CD36 in the liver, resulting in the reduction of body weight and fat volume in HFD-fed obese mice. Notably, we also observed that the administration of at least 106 CFU of L. plantarum LMT1-48 significantly lowered body weight and abdominal fat volume in modified diet-fed mouse models. Collectively, these data suggest that L. plantarum LMT1-48 is a potential healthy food for obese people.


Subject(s)
Anti-Obesity Agents/pharmacology , Lactobacillus plantarum , Lipogenesis/genetics , Obesity/diet therapy , 3T3-L1 Cells , Animals , Diet, High-Fat/adverse effects , Gene Expression Regulation , Leptin/blood , Lipogenesis/physiology , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Obesity/metabolism , Probiotics/pharmacology , Triglycerides/metabolism
2.
J Med Food ; 22(6): 560-566, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31013456

ABSTRACT

The gut microbiota is the most important environmental factor that plays a role in inducing obesity. The gram-negative bacteria, Enterobacter cloacae strains, recently identified in obese mice are considered to be pathogenic bacteria in the gut. Probiotics are important members of the gut microbiota and exert beneficial effects, including inhibiting the growth of potential pathogenic bacteria. Therefore, we isolated a total of 230 lactic acid bacteria from traditional, Korean fermented foods and fecal samples from newborn infants, including Lactobacillus plantarum LMT1-48, which exhibited maximal antimicrobial activity against E. cloacae. We next investigated the functional antiobesity effects of L. plantarum LMT1-48 in an E. cloacae-induced high-fat diet (HFD)-fed animal obesity model. To this end, the L. plantarum LMT1-48 showed antiobesity effects, including body weight loss and reduction of abdominal fat volume, which was accompanied by a decrease in leptin and total cholesterol levels in E. cloacae-induced HFD-fed mice. Notably, gut microbiota diversity also increased after long-term ingestion of L. plantarum LMT1-48, resulting in amelioration of obesity in E. cloacae-induced HFD-fed mice. Accordingly, results suggest that dietary intake of L. plantarum LMT1-48 protects against the onset of E. cloacae-induced obesity.


Subject(s)
Anti-Obesity Agents/administration & dosage , Enterobacter cloacae/physiology , Lactobacillus plantarum/physiology , Obesity/drug therapy , Probiotics/administration & dosage , Animals , Antibiosis , Body Weight/drug effects , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome , Humans , Intestines/microbiology , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Obesity/microbiology , Triglycerides/metabolism
3.
Cell Rep ; 27(3): 699-707.e4, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995469

ABSTRACT

Kelch-like ECH-associated protein 1 (Keap1) and nuclear factor (erythroid-derived 2)-like 2 (Nrf2) proteins work in concert to regulate the levels of reactive oxygen species (ROS). The Keap1-Nrf2 antioxidant system also participates in T cell differentiation and inflammation, but its role in innate T cell development and functions remains unclear. We report that T cell-specific deletion of Keap1 results in defective development and reduced numbers of invariant natural killer T (NKT) cells in the thymus and the peripheral organs in a cell-intrinsic manner. The frequency of NKT2 and NKT17 cells increases while NKT1 decreases in these mice. Keap1-deficient NKT cells show increased rates of proliferation and apoptosis, as well as increased glucose uptake and mitochondrial function, but reduced ROS, CD122, and Bcl2 expression. In NKT cells deficient in Nrf2 and Keap1, all these phenotypic and metabolic defects are corrected. Thus, the Keap1-Nrf2 system contributes to NKT cell development and homeostasis by regulating cell metabolism.


Subject(s)
Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Natural Killer T-Cells/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cell Survival , Glucose/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Interleukin-2 Receptor beta Subunit/genetics , Interleukin-2 Receptor beta Subunit/metabolism , Kelch-Like ECH-Associated Protein 1/deficiency , Kelch-Like ECH-Associated Protein 1/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics , Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Thymus Gland/metabolism
4.
J Clin Invest ; 128(11): 4970-4979, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30106382

ABSTRACT

Graft-versus-host disease (GVHD) in the gastrointestinal (GI) tract remains the major cause of morbidity and nonrelapse mortality after BM transplantation (BMT). The Paneth cell protein regenerating islet-derived 3α (REG3α) is a biomarker specific for GI GVHD. REG3α serum levels rose in the systematic circulation as GVHD progressively destroyed Paneth cells and reduced GI epithelial barrier function. Paradoxically, GVHD suppressed intestinal REG3γ (the mouse homolog of human REG3α), and the absence of REG3γ in BMT recipients intensified GVHD but did not change the composition of the microbiome. IL-22 administration restored REG3γ production and prevented apoptosis of both intestinal stem cells (ISCs) and Paneth cells, but this protection was completely abrogated in Reg3g-/- mice. In vitro, addition of REG3α reduced the apoptosis of colonic cell lines. Strategies that increase intestinal REG3α/γ to promote crypt regeneration may offer a novel, nonimmunosuppressive approach for GVHD and perhaps for other diseases involving the ISC niche, such as inflammatory bowel disease.


Subject(s)
Apoptosis , Bone Marrow Transplantation , Colon/metabolism , Graft vs Host Disease/metabolism , Inflammatory Bowel Diseases/metabolism , Pancreatitis-Associated Proteins/metabolism , Paneth Cells/metabolism , Signal Transduction , Animals , Cell Survival/genetics , Colon/pathology , Female , Graft vs Host Disease/pathology , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Pancreatitis-Associated Proteins/genetics , Paneth Cells/pathology , Prospective Studies , Transplantation, Homologous
5.
Eur J Immunol ; 48(7): 1255-1257, 2018 07.
Article in English | MEDLINE | ID: mdl-29572809

ABSTRACT

We show the presence of lymphoid tissue-resident PLZF+ CD45RA+ RO+ CD4 T cells in humans. They express HLA-DR, granzyme B, and perforin and are low on CCR7 like terminally differentiated effector memory (Temra) cells and are likely generated from effector T cells (Te) or from central (Tcm) or effector (Tem) memory T (Tcm) cells during immune responses. Tn, Naïve T cells.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Genotype , Lymphoid Tissue/immunology , Promyelocytic Leukemia Zinc Finger Protein/metabolism , T-Lymphocyte Subsets/physiology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Cells, Cultured , Granzymes/metabolism , HLA-DR Antigens/metabolism , Humans , Immunity, Cellular , Immunologic Memory , Perforin/metabolism
6.
J Immunol ; 199(10): 3478-3487, 2017 11 15.
Article in English | MEDLINE | ID: mdl-29021374

ABSTRACT

Reactive oxygen species (ROS) are byproducts of aerobic metabolism and contribute to both physiological and pathological conditions as second messengers. ROS are essential for activation of T cells, but how ROS influence NKT cells is unknown. In the present study, we investigated the role of ROS in NKT cell function. We found that NKT cells, but not CD4 or CD8 T cells, have dramatically high ROS in the spleen and liver of mice but not in the thymus or adipose tissues. Accordingly, ROS-high NKT cells exhibited increased susceptibility and apoptotic cell death with oxidative stress. High ROS in the peripheral NKT cells were primarily produced by NADPH oxidases and not mitochondria. We observed that sorted ROS-high NKT cells were enriched in NKT1 and NKT17 cells, whereas NKT2 cells were dominant in ROS-low cells. Furthermore, treatment of NKT cells with antioxidants led to reduced frequencies of IFN-γ- and IL-17-expressing cells, indicating that ROS play a role in regulating the inflammatory function of NKT cells. The transcription factor promyelocytic leukemia zinc finger (PLZF) seemed to control the ROS levels. NKT cells from adipose tissues that do not express PLZF and those from PLZF haplodeficient mice have low ROS. Conversely, ROS were highly elevated in CD4 T cells from mice ectopically expressing PLZF. Thus, our findings demonstrate that PLZF controls ROS levels, which in turn governs the inflammatory function of NKT cells.


Subject(s)
Liver/immunology , Natural Killer T-Cells/immunology , Promyelocytic Leukemia Zinc Finger Protein/metabolism , Reactive Oxygen Species/metabolism , Spleen/immunology , Animals , Apoptosis , Cells, Cultured , Interferon-gamma/metabolism , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NADPH Oxidases/metabolism , Oxidative Stress , Promyelocytic Leukemia Zinc Finger Protein/genetics
7.
J Hepatol ; 67(1): 100-109, 2017 07.
Article in English | MEDLINE | ID: mdl-28267623

ABSTRACT

BACKGROUND & AIMS: The liver is an immunologically-privileged organ. Breakdown of liver immune privilege has been reported in chronic liver disease; however, the role of adaptive immunity in liver injury is poorly defined. Nuclear factor-κB-inducing kinase (NIK) is known to regulate immune tissue development, but its role in maintaining liver homeostasis remains unknown. This study aimed to assess the role of NIK, particularly thymic NIK, in regulating liver adaptive immunity. METHODS: NIK was deleted systemically or conditionally using the Cre/loxp system. Cluster of differentiation [CD]4+ or CD8+ T cells were depleted using anti-CD4 or anti-CD8 antibody. Donor bone marrows or thymi were transferred into recipient mice. Immune cells were assessed by immunohistochemistry and flow cytometry. RESULTS: Global, but not liver-specific or hematopoietic lineage cell-specific, deletion of NIK induced fatal liver injury, inflammation, and fibrosis. Likewise, adoptive transfer of NIK-null, but not wild-type, thymi into immune-deficient mice induced liver inflammation, injury, and fibrosis in recipients. Liver inflammation was characterized by a massive expansion of T cells, particularly the CD4+ T cell subpopulation. Depletion of CD4+, but not CD8+, T cells fully protected against liver injury, inflammation, and fibrosis in NIK-null mice. NIK deficiency also resulted in inflammation in the lung, kidney, and pancreas, but to a lesser degree relative to the liver. CONCLUSIONS: Thymic NIK suppresses development of autoreactive T cells against liver antigens, and NIK deficiency in the thymus results in CD4+ T cell-orchestrated autoimmune hepatitis and liver fibrosis. Thus, thymic NIK is essential for the maintenance of liver immune privilege and liver homeostasis. LAY SUMMARY: We found that global or thymus-specific ablation of the NIK gene results in fatal autoimmune liver disease in mice. NIK-deficient mice develop liver inflammation, injury, and fibrosis. Our findings indicate that thymic NIK is essential for the maintenance of liver integrity and homeostasis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Hepatitis, Autoimmune/etiology , Liver Cirrhosis, Experimental/etiology , Liver/immunology , Protein Serine-Threonine Kinases/physiology , Thymus Gland/physiology , Adaptive Immunity , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappaB-Inducing Kinase
8.
BMC Immunol ; 16: 62, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26482437

ABSTRACT

BACKGROUND: Invariant Natural Killer T (iNKT) cells have been implicated in lung inflammation in humans and also shown to be a key cell type in inducing allergic lung inflammation in mouse models. iNKT cells differentiate and acquire functional characteristics during development in the thymus. However, the correlation between development of iNKT cells in the thymus and role in lung inflammation remains unknown. In addition, transcriptional control of differentiation of iNKT cells into iNKT cell effector subsets in the thymus during development is also unclear. In this report we show that ß-catenin dependent mechanisms direct differentiation of iNKT2 and iNKT17 subsets but not iNKT1 cells. METHODS: To study the role for ß-catenin in lung inflammation we utilize mice with conditional deletion and enforced expression of ß-catenin in a well-established mouse model for IL-25-dependen lung inflammation. RESULTS: Specifically, we demonstrate that conditional deletion of ß-catenin permitted development of mature iNKT1 cells while impeding maturation of iNKT2 and 17 cells. A role for ß-catenin expression in promoting iNKT2 and iNKT17 subsets was confirmed when we noted that enforced transgenic expression of ß-catenin in iNKT cell precursors enhanced the frequency and number of iNKT2 and iNKT17 cells at the cost of iNKT1 cells. This effect of expression of ß-catenin in iNKT cell precursors was cell autonomous. Furthermore, iNKT2 cells acquired greater capability to produce type-2 cytokines when ß-catenin expression was enhanced. DISCUSSION: This report shows that ß-catenin deficiency resulted in a profound decrease in iNKT2 and iNKT17 subsets of iNKT cells whereas iNKT1 cells developed normally. By contrast, enforced expression of ß-catenin promoted the development of iNKT2 and iNKT17 cells. It was important to note that the majority of iNKT cells in the thymus of C57BL/6 mice were iNKT1 cells and enforced expression of ß-catenin altered the pattern to iNKT2 and iNKT17 cells suggesting that ß-catenin may be a major factor in the distinct pathways that critically direct differentiation of iNKT effector subsets. CONCLUSIONS: Thus, we demonstrate that ß-catenin expression in iNKT cell precursors promotes differentiation toward iNKT2 and iNKT17 effector subsets and supports enhanced capacity to produce type 2 and 17 cytokines which in turn augment lung inflammation in mice.


Subject(s)
Cell Differentiation , Interleukin-17/metabolism , Natural Killer T-Cells/immunology , Pneumonia/immunology , Pneumonia/pathology , beta Catenin/metabolism , Animals , Bronchial Hyperreactivity/complications , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/pathology , Mice, Inbred C57BL , Mice, Transgenic , Pneumonia/complications
9.
Biol Blood Marrow Transplant ; 19(8): 1244-53, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23756210

ABSTRACT

The effect of minor H antigen mismatching on the occurrence of graft-versus-host disease (GvHD) and graft-versus-leukemia (GvL) after HLA-matched hematopoietic stem cell transplantation (HSCT) has mainly been demonstrated in single-center studies. Yet, the International Histocompatibility and Immunogenetics Workshops (IHIW) provide a collaborative platform to execute crucial large studies. In collaboration with 20 laboratories of the IHIW, the roles of 10 autosomal and 10 Y chromosome-encoded minor H antigens were investigated on GvHD and relapse incidence in 639 HLA-identical related donor (IRD) and 210 HLA-matched unrelated donor (MUD) HSCT recipients. Donor and recipient DNA samples were genotyped for the minor H antigens HA-1, HA-2, HA-3, HA-8, HB-1, ACC-1, ACC-2, SP110, PANE1, UGT2B17, and HY. The correlations with the primary outcomes GvHD (acute or chronic GvHD), survival, and relapse were statistically analyzed. The results of these multicenter analyses show that none of the HLA class I-restricted HY antigens were found to be associated with any of the primary outcomes. Interestingly, of the HLA class II-restricted HY antigens analyzed, HLA-DQ5 positive recipients showed a significantly increased GvHD-free survival in female-to-male HSCT compared with male-to-female HSCT (P = .013). Yet, analysis of the overall gender effect, thus independent of the known HY antigens, between the gender groups demonstrated an increased GvHD incidence in the female-to-male transplantations (P < .005) and a decreased GvHD-free survival in the female-to-male transplantations (P < .001). Of all autosomally encoded minor H antigens, only mismatching for the broadly expressed minor H antigen HA-8 increased the GvHD incidence in IRD HSCT (Hazard ratio [HR] = 5.28, P < .005), but not in MUD HSCT. Most striking was the influence of hematopoietic restricted minor H antigens on GvL as mismatching for hematopoietic minor H antigens correlated with lower relapse rates (P = .078), higher relapse-free survival (P = .029), and higher overall survival (P = .032) in recipients with GvHD, but not in those without GvHD. In conclusion, the significant GvHD effect of the broadly expressed minor H antigen HA-8 favors matching for HA-8 in IRD, but not in MUD, patient/donor pairs. The GvHD-GvL association demonstrating a significant lower relapse in hematopoietic minor H antigen mismatched patient/donor pairs underlines their clinical applicability for adoptive immunotherapy, enhancing the GvL effect in a GvHD controllable manner.


Subject(s)
Graft vs Host Disease/immunology , Graft vs Leukemia Effect/immunology , Hematopoietic Stem Cell Transplantation/methods , Histocompatibility/immunology , Minor Histocompatibility Antigens/immunology , Adult , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Histocompatibility Testing , Humans , Male , Unrelated Donors
10.
Biol Blood Marrow Transplant ; 18(3): 381-7, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22062805

ABSTRACT

HY-specific T cells are presumed to play a role in acute graft-versus-host disease (aGVHD) after female-to-male stem cell transplantation (SCT). However, infiltrates of these T cells in aGVHD-affected tissues have not yet been reported. We evaluated the application of HLA-A2/HY dextramers for the in situ detection of HY-specific T cells in cryopreserved skin biopsy specimens. We applied the HLA-A2/HY dextramers on cryopreserved skin biopsy specimens from seven male HLA-A2(+) pediatric patients who underwent stem cell transplantation with confirmed aGVHD involving the skin. The dextramers demonstrated the presence of HY-specific T cells. In skin biopsy specimens of three male recipients of female grafts, 68% to 78% of all skin-infiltrating CD8(+) T cells were HY-specific, whereas these cells were absent in biopsy specimens collected from sex-matched patient-donor pairs. Although this study involved a small and heterogeneous patient group, our results strongly support the hypothesis that HY-specific T cells are actively involved in the pathophysiology of aGVHD after sex-mismatched stem cell transplantation.


Subject(s)
Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/methods , Skin/immunology , T-Lymphocytes/immunology , Adolescent , Child , Child, Preschool , Cryopreservation , Fluorescent Antibody Technique , Graft vs Host Disease/pathology , Humans , Male , Microscopy, Confocal , Skin/pathology , T-Lymphocytes/pathology
11.
Biol Blood Marrow Transplant ; 17(1): 69-77, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20708701

ABSTRACT

T cells specific for hematopoietic system restricted minor Histocompatibility (H) antigens target normal and malignant hematopoietic cells. Thus, cellular immune responses against the latter miHAS eradicate the recipient's hematopoiesis including residual leukemic cells after HLA-matched minor H antigen-mismatched stem-cell transplantation (SCT). However, there are controversial reports on the role of HA-1 in the development of graft-versus-host-disease (GVHD) as well. Here, we address the behavior of HA-1-specific cytotoxic T cells (CTLs) in an ex vivo in situ skin explant model wherein HA-1-expressing dendritic cells (DCs) were added as antigen-presenting cells (APCs). Infiltration and activation of HA-1 CTLs occurred only in those cases where both HLA-A2 and HA-1 were expressed, either by the skin or by the DCs, or by the combination of HLA-A2(+) skin and HA-1(+) DCs. These results point toward the role of recipient's HA-1(+) DCs in the chimeric patient suffering from GVHD after HA-1-mismatched SCT. Although in our model the infiltrated and activated CTLs did not cause skin tissue destruction, our results provide a first step in understanding the reported association of HA-1 mismatching with clinical GVHD.


Subject(s)
Dendritic Cells/transplantation , Graft vs Host Disease/therapy , Immunotherapy/methods , Minor Histocompatibility Antigens , Oligopeptides , T-Lymphocytes, Cytotoxic/immunology , Cell Movement , Dendritic Cells/immunology , Humans , In Vitro Techniques , Lymphocyte Activation , Models, Biological , Skin/cytology , Skin/immunology , Skin Diseases/therapy
12.
J Immunol Methods ; 310(1-2): 78-85, 2006 Mar 20.
Article in English | MEDLINE | ID: mdl-16487538

ABSTRACT

Tetrameric MHC/peptide complexes are important tools for analyzing antigen-specific T cells. The in situ use of tetrameric MHC/peptide complexes in viable tissue sections has several shortcomings: it does not allow the execution of multiple analyses on one single biopsy, the storage of the biopsies, and the co-staining of the tetramer-positive cells for various intracellular molecules. We have developed a novel approach using overnight pre-labeling of viable human tissues with MHC/peptide tetramers, followed by cryopreservation and labeling of the cryosections. The visualization of antigen-specific T cells, combined with detection of other membrane, cytoplasmic, or nuclear markers is now feasible.


Subject(s)
Skin/immunology , T-Lymphocytes/immunology , Biopsy , Cryopreservation , Female , Fluorescent Dyes/chemistry , H-Y Antigen/analysis , H-Y Antigen/immunology , HLA-A2 Antigen/analysis , HLA-A2 Antigen/immunology , Humans , Immunohistochemistry , In Vitro Techniques , Male , Skin/cytology , T-Lymphocytes/cytology
13.
Infect Immun ; 72(4): 2014-21, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15039321

ABSTRACT

We report here the induction of specific protective cellular immunity against Mycobacterium tuberculosis by the employment of vaccination with recombinant attenuated Listeria monocytogenes strains. We constructed self-destructing attenuated L. monocytogenes Delta 2 strains carrying eukaryotic expression plasmids for the antigen 85 complex (Ag85A and Ag85B) and for MPB/MPT51 (mycobacterial protein secreted by M. bovis BCG/mycobacterial protein secreted by M. tuberculosis) molecules. Infection of these recombinant bacteria allowed expression of the genes in the J774A.1 murine macrophage cell line. Intraperitoneal vaccination of C57BL/6 mice with these recombinant bacteria was capable of inducing purified protein derivative-specific cellular immune responses, such as foot pad reactions, proliferative responses of splenocytes, and gamma interferon production from splenocytes, suggesting the efficacy of vaccination against mycobacterial infection by use of these recombinant L. monocytogenes strains. Furthermore, intravenous vaccination with recombinant bacteria carrying expression plasmids for Ag85A, Ag85B, or MPB/MPT51 in BALB/c mice elicited significant protective responses, comparable to those evoked by a live Mycobacterium bovis BCG vaccine. Notably, this is the first report to show that MPB/MPT51 is a major protective antigen in addition to Ag85A and Ag85B, which have been reported to be major mycobacterial protective antigens.


Subject(s)
Immunity, Cellular , Listeria monocytogenes/genetics , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/immunology , Vaccines, Attenuated/immunology , Vaccines, DNA/immunology , Acyltransferases/genetics , Animals , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Cattle , Cell Line , Immunization , Macrophages , Mice , Mice, Inbred BALB C , Plasmids/genetics , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/genetics , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/prevention & control , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, DNA/administration & dosage
14.
Naunyn Schmiedebergs Arch Pharmacol ; 369(4): 402-7, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14999439

ABSTRACT

Stress proteins have been implicated in pathological cardiovascular conditions. We hypothesized that a heat-shock response modulates contractility of vascular smooth muscles. Rat aortic ring preparations were mounted in organ baths, exposed to 42 degrees C for 45 min, and subjected to contractions. Expression of HSP70 and phosphorylation of myosin light chain were examined with immunoblots. Heat shock enhanced contractile response to KCl in parallel with HSP70 expression in rat aortic rings from 8 h but not 1 h after the end of heat shock. Heat shock also augmented vascular contractility to phenylephrine whether endothelium was intact or denuded. Treatment of heat shock-preconditioned aortic rings with Bay K8644, a calcium channel activator, but not treatment with phorbol dibutyrate (1 micromol/l), a protein kinase C activator, enhanced contractions of the rings as compared with those of the control. The levels of phosphorylation of myosin light chains after administration of phenylephrine in heat shock-preconditioned tissues were statistically significantly higher than those in control tissues. Pretreatment with wortmannin (300 nmol/l), an inhibitor of myosin light chain kinase, decreased both contractility and phosphorylation of myosin light chains in parallel. However, heat-shock response did not affect relaxation responses to either acetylcholine in endothelium-intact aortic rings or sodium nitroprusside in endothelium-denuded rings. These results suggest that the heat-shock response is associated with enhanced vascular smooth muscle contractility through a modulation of thick-filament regulation.


Subject(s)
Aorta, Thoracic/physiology , Endothelium, Vascular/physiology , Heat-Shock Response/physiology , Muscle, Smooth, Vascular/physiology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Androstadienes/pharmacology , Animals , Aorta, Thoracic/drug effects , Calcium/metabolism , Calcium Channel Agonists/pharmacology , Endothelium, Vascular/drug effects , HSP70 Heat-Shock Proteins/biosynthesis , Heat-Shock Response/drug effects , Immunoblotting , In Vitro Techniques , Male , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth, Vascular/drug effects , Myosin Light Chains/biosynthesis , Myosin-Light-Chain Kinase/antagonists & inhibitors , Phenylephrine/pharmacology , Phosphorylation , Potassium Chloride/pharmacology , Rats , Rats, Sprague-Dawley , Vasoconstrictor Agents/pharmacology , Wortmannin
15.
Infect Immun ; 70(5): 2676-80, 2002 May.
Article in English | MEDLINE | ID: mdl-11953411

ABSTRACT

Listeria epitope-specific helper T (Th) cells were able to be primed and induced in vivo by immunization with a plasmid carrying an invariant chain (Ii) gene whose class II-associated invariant chain peptide (CLIP) region was replaced by a Listeria Th epitope. Immunization of C3H/He mice with an Ii-LLO 215-226 plasmid induced specific interferon-gamma- and interleukin 2-producing Th cells and conferred significant protective immunity against listerial infection.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/genetics , Bacterial Toxins , Bacterial Vaccines/immunology , Epitopes, T-Lymphocyte , Heat-Shock Proteins/genetics , Histocompatibility Antigens Class II/genetics , Listeria monocytogenes/immunology , T-Lymphocytes, Helper-Inducer/immunology , Vaccines, DNA/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/immunology , Heat-Shock Proteins/immunology , Hemolysin Proteins , Histocompatibility Antigens Class II/immunology , Immunization , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Mice , Mice, Inbred C3H , Peptide Fragments/immunology , Plasmids
SELECTION OF CITATIONS
SEARCH DETAIL
...