Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Immunity ; 54(4): 721-736.e10, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33725478

ABSTRACT

Hyperglycemia and hyperlipidemia are often observed in individuals with type II diabetes (T2D) and related mouse models. One dysmetabolic biochemical consequence is the non-enzymatic reaction between sugars, lipids, and proteins, favoring protein glycation, glycoxidation, and lipoxidation. Here, we identified oxidative alterations in key components of the major histocompatibility complex (MHC) class II molecule antigen processing and presentation machinery in vivo under conditions of hyperglycemia-induced metabolic stress. These modifications were linked to epitope-specific changes in endosomal processing efficiency, MHC class II-peptide binding, and DM editing activity. Moreover, we observed some quantitative and qualitative changes in the MHC class II immunopeptidome of Ob/Ob mice on a high-fat diet compared with controls, including changes in the presentation of an apolipoprotein B100 peptide associated previously with T2D and metabolic syndrome-related clinical complications. These findings highlight a link between glycation reactions and altered MHC class II antigen presentation that may contribute to T2D complications.


Subject(s)
Antigen Presentation/immunology , Histocompatibility Antigens Class II/immunology , Stress, Physiological/immunology , Animals , Antigen-Presenting Cells/immunology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 2/immunology , Disease Models, Animal , Epitopes/immunology , Female , Male , Mice , Mice, Inbred C57BL , Peptides/immunology , Protein Binding/immunology
2.
J Immunol ; 206(3): 494-504, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33318291

ABSTRACT

The expression and turnover of Ag-specific peptide-MHC class II (pMHC-II) on the surface of dendritic cells (DCs) is essential for their ability to efficiently activate CD4 T cells. Ubiquitination of pMHC-II by the E3 ubiquitin ligase March-I regulates surface expression and survival of pMHC-II in DCs. We now show that despite their high levels of surface pMHC-II, MHC class II (MHC-II) ubiquitination-deficient mouse DCs are functionally defective; they are poor stimulators of naive CD4 T cells and secrete IL-12 in response to LPS stimulation poorly. MHC-II ubiquitination-mutant DC defects are cell intrinsic, and single-cell RNA sequencing demonstrates that these DCs have an altered gene expression signature as compared with wild-type DCs. Curiously, these functional and gene transcription defects are reversed by activating the DCs with LPS. These results show that dysregulation of MHC-II turnover suppresses DC development and function.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Ubiquitin-Protein Ligases/metabolism , Animals , Antigen Presentation , Antigens/metabolism , Cell Differentiation , Cells, Cultured , Histocompatibility Antigens Class II/metabolism , Interleukin-12/metabolism , Lymphocyte Activation/genetics , Mice , Mice, Knockout , Ubiquitin-Protein Ligases/genetics , Ubiquitination
3.
J Biol Chem ; 290(20): 12804-11, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25847238

ABSTRACT

Serum amyloid A is a proinflammatory molecule that induces leukocyte infiltration and promotes neutrophil adhesion to endothelial cells under inflammatory conditions. The aim of this study was to examine whether Saa1 aggravates T cell-mediated hepatitis by inducing chemokines in a liver-specific, Saa1-overexpressing, transgenic (TG) mouse model. We generated TG mice in which Saa1 was overexpressed specifically in liver tissue. The chemokines monocyte chemotactic protein 1 (MCP1), MIP1α, MIP1ß, interferon γ-induced protein 10 (IP-10), and eotaxin were induced in Saa1 TG mice. After concanavalin A treatment, Saa1 expression was higher in Saa1 TG mice than in WT mice. More severe liver injury, increased hepatocyte apoptosis, and higher levels of hepatic enzymes were observed in Saa1 TG mice than in WT mice. Liver infiltration of CD4(+) T cells and macrophages increased after inducing hepatitis. Activation of T cells was higher in Saa1 TG mice than in WT mice, and the populations of Th17 cells and regulatory T cells were altered by overexpressing Saa1 in TG mice. Secretion of various cytokines, such as interferon γ, tumor necrosis factor α, and interleukin 6, increased in Saa1 TG mice. Injecting a Toll-like receptor 2 (TLR2) antagonist in vivo inhibited chemokine expression and IκBα phosphorylation and showed that the induction of chemokines by Saa1 was dependent on TLR2. Hepatic Saa1 accelerated T cell-mediated hepatitis by inducing chemokine production and activating T cells by TLR2. Therefore, Saa1 might be a novel inflammatory factor that acts as a chemokine modulator in hepatitis.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Chemokines/biosynthesis , Inflammation Mediators/metabolism , Serum Amyloid A Protein/biosynthesis , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism , Toll-Like Receptor 2/metabolism , Animals , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Chemokines/genetics , Concanavalin A/adverse effects , Concanavalin A/pharmacology , Liver/metabolism , Liver/pathology , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Transgenic , Mitogens/adverse effects , Mitogens/pharmacology , Serum Amyloid A Protein/genetics , T-Lymphocytes, Regulatory/pathology , Th17 Cells/pathology , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 2/genetics
4.
Differentiation ; 89(1-2): 42-50, 2015.
Article in English | MEDLINE | ID: mdl-25766428

ABSTRACT

Mouse embryonic stem cells (ESCs) are self-renewing, pluripotent, and have the ability to differentiate into the three germ layers required to form all embryonic tissues. These properties are maintained by both intrinsic and extrinsic factors. Many studies have contributed to the understanding of the molecular signal transduction required for pluripotency and controlled differentiation. Such an understanding is important in the potential application of stem cells to cell therapy for disease, and thus there is an interest in understanding the cell cycle regulation, pluripotency, and differentiation of ESCs. The regulator of G protein signaling (RGS) family consists of over 20 members. Rgs19, one such protein, specifically interacts with Gαi to enhance its GTPase activity. Growth factor receptors use Gi proteins for signal transduction, and Rgs19 may thus be involved in the regulation of cell proliferation. In a previous gain-of-function study, Rgs19 overexpression was found to enhance proliferation in various cell types. Our data demonstrate a role for Rgs19 in the regulation of ESC differentiation. Based on the presence of Rgs19 in ESCs, the morphological and molecular properties of wild-type and Rgs19 +/- ESCs during LIF withdrawal, in vitro differentiation, and teratoma formation were compared. Our findings provide insight for the first time into the mechanisms involved in Rgs19 regulation of mouse ESC proliferation and differentiation.


Subject(s)
Cell Differentiation/genetics , Cell Proliferation/genetics , Mouse Embryonic Stem Cells , RGS Proteins/genetics , Animals , Gene Expression Regulation, Developmental , Mice , RGS Proteins/biosynthesis , Signal Transduction
5.
Arterioscler Thromb Vasc Biol ; 34(10): 2276-82, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25147341

ABSTRACT

OBJECTIVE: Angiogenesis is an important biological process during development, reproduction, and in immune responses. Placental growth factor (PlGF) is a member of vascular endothelial growth factor that is critical for angiogenesis and vasculogenesis. We generated transgenic mice overexpressing PlGF in specifically T cells using the human CD2-promoter to investigate the effects of PlGF overexpression. APPROACH AND RESULTS: Transgenic mice were difficult to obtain owing to high lethality; for this reason, we investigated why gestational loss occurred in these transgenic mice. Here, we report that placenta detachment and inhibition of angiogenesis occurred in PlGF transgenic mice during the gestational period. Moreover, even when transgenic mice were born, their growth was restricted. CONCLUSIONS: Conclusively, PlGF overexpression prevents angiogenesis by inhibiting Braf, extracellular signal-regulated kinase activation, and downregulation of HIF-1α in the mouse placenta. Furthermore, it affected regulatory T cells, which are important for maintenance of pregnancy.


Subject(s)
Fetal Death/metabolism , Fetal Growth Retardation/metabolism , Neovascularization, Physiologic , Placenta/blood supply , Placenta/metabolism , Pregnancy Proteins/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Body Weight , CD2 Antigens/genetics , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fetal Death/genetics , Fetal Death/physiopathology , Fetal Growth Retardation/genetics , Fetal Growth Retardation/physiopathology , Gestational Age , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Litter Size , Mice , Mice, Inbred C57BL , Mice, Transgenic , Placenta Growth Factor , Pregnancy , Pregnancy Proteins/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction , Up-Regulation
6.
Biochem Biophys Res Commun ; 452(3): 822-7, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25201726

ABSTRACT

Chronic hepatitis is a major cause of liver cancer, so earlier treatment of hepatitis might be reducing liver cancer incidence. Hepatitis can be induced in mice by treatment with Concanavalin A (Con A); the resulting liver injury causes significant CD4(+) T cell activation and infiltration. In these T cells, Roquin, a ring-type E3 ubiquitin ligase, is activated. To investigate the role of Roquin, we examined Con A-induced liver injury and T cell infiltration in transgenic (Tg) mice overexpressing Roquin specifically in T cells. In Roquin Tg mice, Con A treatment caused greater increases in both the levels of liver injury enzymes and liver tissue apoptosis, as revealed by TUNEL and H&E staining, than wild type (WT) mice. Further, Roquin Tg mice respond to Con A treatment with greater increases in the T cell population, particularly Th17 cells, though Treg cell counts are lower. Roquin overexpression also enhances increases in pro-inflammatory cytokines, including IFN-γ, TNF-α and IL-6, upon liver injury. Furthermore, Roquin regulates the immune response and apoptosis in Con A induced hepatitis via STATs, Bax and Bcl2. These findings suggest that over-expression of Roquin exacerbates T-cell mediated hepatitis.


Subject(s)
Chemical and Drug Induced Liver Injury/genetics , Hepatocytes/metabolism , Promoter Regions, Genetic , Th17 Cells/metabolism , Ubiquitin-Protein Ligases/genetics , Animals , Apoptosis , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Concanavalin A , Female , Gene Expression Regulation , Hepatocytes/pathology , Interferon-gamma/biosynthesis , Interferon-gamma/metabolism , Interleukin-6/biosynthesis , Interleukin-6/metabolism , Lymphocyte Activation , Lymphocyte Count , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Th17 Cells/pathology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin-Protein Ligases/metabolism
7.
Biochem Biophys Res Commun ; 444(3): 296-301, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24380856

ABSTRACT

Jazf1 is a 27 kDa nuclear protein containing three putative zinc finger motifs that is associated with diabetes mellitus and prostate cancer; however, little is known about the role that this gene plays in regulation of metabolism. Recent evidence indicates that Jazf1 transcription factors bind to the nuclear orphan receptor TR4. This receptor regulates PEPCK, the key enzyme involved in gluconeogenesis. To elucidate Jazf1's role in metabolism, we fed a 60% fat diet for up to 15 weeks. In Jazf1 overexpression mice, weight gain was found to be significantly decreased. The expression of Jazf1 in the liver also suppressed lipid accumulation and decreased droplet size. These results suggest that Jazf1 plays a critical role in the regulation of lipid homeostasis. Finally, Jazf1 may provide a new therapeutic target in the management of obesity and diabetes.


Subject(s)
Carrier Proteins/genetics , Diet, High-Fat , Lipid Metabolism/genetics , Nuclear Proteins/genetics , Weight Gain/genetics , Animals , Blood Glucose/analysis , Co-Repressor Proteins , DNA-Binding Proteins , Glucose Tolerance Test , Homeostasis , Insulin/physiology , Mice , Mice, Transgenic , Phosphoenolpyruvate Carboxykinase (GTP)/antagonists & inhibitors , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
8.
J Biol Chem ; 287(50): 42269-77, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23066015

ABSTRACT

To investigate the role of Roquin, a RING-type ubiquitin ligase family member, we used transgenic mice with enforced Roquin expression in T cells, with collagen-induced arthritis (CIA). Wild-type (WT) and Roquin transgenic (Tg) mice were immunized with bovine type II collagen (CII). Arthritis severity was evaluated by clinical score; histopathologic CIA severity; proinflammatory and anti-inflammatory cytokine levels; anti-CII antibody levels; and populations of Th1, Th2, germinal center B cells, and follicular helper T cells in CIA. T cell proliferation in vitro and cytokine levels were determined to assess the response to CII. Roquin Tg mice developed more severe CIA and joint destruction compared with WT mice. Production of TNF-α, IFN-γ, IL-6, and pathogenic anti-collagen CII-specific IgG and IgG2a antibodies was increased in Roquin Tg mice. In addition, in vitro T cell assays showed increased proliferation and proinflammatory cytokine production in response to CII as a result of enforced Roquin expression in T cells. Furthermore, the Th1/Th2 balance was altered by an increased Th1 and decreased Th2 population. These findings suggest that overexpression of Roquin exacerbates the development of CIA and that enforced expression of Roquin in T cells may promote autoimmune diseases such as CIA.


Subject(s)
Arthritis, Experimental/immunology , Cell Proliferation , Gene Expression Regulation/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Ubiquitin-Protein Ligases/immunology , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cattle , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/immunology , Gene Expression Regulation/genetics , Germinal Center/immunology , Germinal Center/metabolism , Germinal Center/pathology , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Mice , Mice, Transgenic , Severity of Illness Index , Th1 Cells/metabolism , Th1 Cells/pathology , Th2 Cells/metabolism , Th2 Cells/pathology , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
9.
Arthritis Rheum ; 64(7): 2191-200, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22275266

ABSTRACT

OBJECTIVE: Calcineurin-binding protein 1 (CABIN-1) regulates calcineurin phosphatase activity as well as the activation, apoptosis, and inflammatory responses of fibroblast-like synoviocytes (FLS), which actively participate in the chronic inflammatory responses in rheumatoid arthritis (RA). However, the mechanism of action of CABIN-1 in FLS apoptosis is not clear. This study was undertaken to define the regulatory role of CABIN-1 in FLS from mice with collagen-induced arthritis (CIA). METHODS: Transgenic mice overexpressing human CABIN-1 in joint tissue under the control of a type II collagen promoter were generated. Expression of human CABIN-1 (hCABIN-1) in joints and FLS was determined by reverse transcription-polymerase chain reaction (RT-PCR) and Western blot analysis. The expression of cytokines, matrix metalloproteinases (MMPs), and apoptosis-related genes in FLS was determined by enzyme-linked immunosorbent assay, gelatin zymography, and RT-PCR, respectively. Joints were stained with hematoxylin and eosin and with tartrate-resistant acid phosphatase for histologic analysis. RESULTS: Human CABIN-1-transgenic mice with CIA had less severe arthritis than wild-type mice with CIA, as assessed according to hind paw thickness and histologic features. The milder arthritis was accompanied by significantly enhanced apoptosis in transgenic mice, evidenced by a significantly greater number of TUNEL-positive cells in synovial tissue. Expression of inflammatory cytokines and MMPs in the transgenic mice with CIA was reduced, and they exhibited decreased Akt activation and increased expression of p53, caspase 3, caspase 9, and Bax. CONCLUSION: Our findings demonstrate that hCABIN-1 plays a critical role in promoting apoptosis of FLS and in attenuating inflammation and cartilage and bone destruction in RA. These results help elucidate the pathogenic mechanisms of RA and suggest that CABIN-1 is a potential target for treatment of this disease.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/physiology , Arthritis, Experimental/pathology , Joints/pathology , Synovial Membrane/pathology , Animals , Arthritis, Experimental/metabolism , Inflammation/metabolism , Inflammation/pathology , Joints/metabolism , Matrix Metalloproteinases/metabolism , Mice , Mice, Transgenic , Synovial Membrane/metabolism
10.
Biochem Biophys Res Commun ; 417(1): 280-6, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22155229

ABSTRACT

The T-cell receptor (TCR) engages with an antigen and initiates a signaling cascade that leads to the activation of transcription factors. Roquin, a protein encoded by the RC3H1 gene and characterized as an immune regulator, was recently identified as a novel RING-type ubiquitin ligase family member, but the mechanisms by which Roquin regulates T-cell responses are unclear. We used the EL-4 murine lymphoma cell line to elucidate the role of Roquin in vitro. Roquin-overexpressing EL-4 cells became hyper-responsive after anti-CD3/CD28 stimulation in vitro and were a major source of the cytokines IL-2 and TNF-α. Upon activation, these cells showed particularly enhanced production of IL-2 and TNF-α. To clarify the important role played by Roquin in T-cell responses ex vivo, we generated T-cell-specific Roquin transgenic (Tg) mice. Roquin-Tg CD4(+) T-cells showed enhanced production of IL-2 and TNF-α in response to TCR stimulation with anti-CD28 co-stimulation. Further studies are necessary to investigate the role of Roquin in the regulation of primary T-cell activation, survival, and differentiation.


Subject(s)
Cytokines/metabolism , Lymphocyte Activation , T-Lymphocytes/immunology , Ubiquitin-Protein Ligases/biosynthesis , Animals , CD28 Antigens/immunology , CD3 Complex/immunology , Cell Line, Tumor , MAP Kinase Kinase 4/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/genetics
11.
Arch Pharm Res ; 45(6): 401-416, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35759090

ABSTRACT

Cancer creates a complex tumor microenvironment (TME) composed of immune cells, stromal cells, blood vessels, and various other cellular and extracellular elements. It is essential for the development of anti-cancer combination therapies to understand and overcome this high heterogeneity and complexity as well as the dynamic interactions between them within the TME. Recent treatment strategies incorporating immune-checkpoint inhibitors and anti-angiogenic agents have brought many changes and advances in clinical cancer treatment. However, there are still challenges for immune suppressive tumors, which are characterized by a lack of T cell infiltration and treatment resistance. In this review, we will investigate the crosstalk between immunity and angiogenesis in the TME. In addition, we will look at strategies designed to enhance anti-cancer immunity, to convert "immune suppressive tumors" into "immune activating tumors," and the mechanisms by which these strategies enhance effector immune cell infiltration.


Subject(s)
Neoplasms , Tumor Microenvironment , Humans , Neoplasms/pathology , Tumor Microenvironment/physiology
12.
J Biol Chem ; 285(37): 28627-34, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20562099

ABSTRACT

Wnt/Wg genes play a critical role in the development of various organisms. For example, the Wnt/beta-catenin signal promotes heart formation and cardiomyocyte differentiation in mice. Previous studies have shown that RGS19 (regulator of G protein signaling 19), which has Galpha subunits with GTPase activity, inhibits the Wnt/beta-catenin signal through inactivation of Galpha(o). In the present study, the effects of RGS19 on mouse cardiac development were observed. In P19 teratocarcinoma cells with RGS19 overexpression, RGS19 inhibited cardiomyocyte differentiation by blocking the Wnt signal. Additionally, several genes targeted by Wnt were down-regulated. For the in vivo study, we generated RGS19-overexpressing transgenic (RGS19 TG) mice. In these transgenic mice, septal defects and thin-walled ventricles were observed during the embryonic phase of development, and the expression of cardiogenesis-related genes, BMP4 and Mef2C, was reduced significantly. RGS19 TG mice showed increased expression levels of brain natriuretic peptide and beta-MHC, which are markers of heart failure, increase of cell proliferation, and electrocardiogram analysis shows abnormal ventricle repolarization. These data provide in vitro and in vivo evidence that RGS19 influenced cardiac development and had negative effects on heart function.


Subject(s)
Cell Differentiation , Heart/embryology , Myocytes, Cardiac/metabolism , RGS Proteins/metabolism , Signal Transduction , Animals , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Cell Line, Tumor , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , Heart Failure/genetics , Heart Failure/metabolism , Heart Septal Defects/genetics , Heart Septal Defects/metabolism , MEF2 Transcription Factors , Mice , Mice, Transgenic , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , RGS Proteins/genetics , Wnt Proteins/genetics , Wnt Proteins/metabolism
13.
Transgenic Res ; 20(5): 1019-31, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21221781

ABSTRACT

The transcription factor Juxtaposed with another zinc finger gene 1 (JAZF1) is a zinc finger protein that binds to the nuclear orphan receptor TR4. Recent evidence indicates that TR4 receptor functions as both a positive and negative regulator of transcription, but the role of JAZF1 in transcriptional mechanisms has not been elucidated. Recently, the incidence rate of congenital heart malformations was reported to be significantly elevated in patients who had neurofibromatosis 1 (NF1) with chromosomal microdeletion syndrome. Furthermore, Joined to JAZF1 (SUZ12) is expressed at high levels in the hearts of adult patients with NF1 microdeletion syndrome. Therefore, we hypothesized that ectopic expression of JAZF1 may lead to cardiac malformations that deleteriously affect the survival of neonates and adults. We sought to elucidate the role of JAZF1 in cardiac development using a Jazf1-overexpressing (Jazf1-Tg) mouse model. In Jazf1-Tg mice, Jazf1 mRNA expression was significantly elevated in the heart. Jazf1-Tg mice also showed cardiac defects, such as high blood pressure, electrocardiogram abnormalities, apoptosis of cardiomyocytes, ventricular non-compaction, and mitochondrial defects. In addition, we found that the expression levels of pro-apoptotic genes were elevated in the hearts of Jazf1-Tg mice. These findings suggest that Jazf1 overexpression may induce heart failure symptoms through the upregulation of pro-apoptotic genes in cardiomyocytes.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Heart Defects, Congenital/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Blood Pressure , Co-Repressor Proteins , DNA-Binding Proteins , Disease Models, Animal , Electrocardiography , Gene Expression Regulation, Developmental , Heart/embryology , Heart/growth & development , Heart Failure/genetics , Mice , Mice, Transgenic , Polycomb Repressive Complex 2 , RNA, Messenger/metabolism , Repressor Proteins/genetics
14.
Biochem Biophys Res Commun ; 374(1): 17-21, 2008 Sep 12.
Article in English | MEDLINE | ID: mdl-18586001

ABSTRACT

The circling (cir/cir) mouse is one of the murine models for human non-syndromic deafness DFNB6. The mice have abnormal circling behavior, suggesting a balanced disorder and profound deafness. The causative gene was transmembrane inner ear (tmie) gene of which the mutation is a 40-kb genomic deletion including tmie gene itself. In this study, tmie-overexpression trasngenic mice were established. Individuals with germline transmission have been mated with circling homozygous mutant mice (cir/cir) in order to produce the transgenic mutant mice (cir/cir-tg) as a gene therapy. After the genotyping, phenotypic analyses were performed so that the insertion of the new gene might compensate for the diseases such as hearing loss, circling behavior, or swimming inability. Some individuals exhibited complete recovery in their behavior and hearing but the others did not show any amelioration in behavior or hearing. Individual mice had very different levels of tmie transgene expression in the cochlea. These results clearly indicate that tmie protein plays an important role when the appropriate expression level of tmie was expressed in the inner ear. The protein levels were variable in each individual and these are thought to induce the differences in disease amelioration levels.


Subject(s)
Genetic Therapy , Hair Cells, Auditory/metabolism , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/therapy , Hearing/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Animals , Disease Models, Animal , Genotype , Hearing Tests , Male , Mice , Mice, Transgenic , Mutation , Phenotype , Transgenes
15.
Cell Transplant ; 17(12): 1371-80, 2008.
Article in English | MEDLINE | ID: mdl-19364074

ABSTRACT

Rheumatoid arthritis is a chronic inflammatory disease. The generation of reactive oxygen species (ROS) within an inflamed joint has been suggested as playing a significant pathogenic role. Extracellular superoxide dismutase (EC-SOD) is a major scavenger enzyme of ROS, which has received growing attention for its therapeutic potential. To investigate the therapeutic effect of EC-SOD in mice with collagen-induced arthritis (CIA), we used mouse embryonic fibroblast (MEF) of transgenic mice that overexpresses EC-SOD on the skin by using hK14 promoter. DBA/1 mice that had been treated with bovine type II collagen were administrated subcutaneous injections of EC-SOD transgenic MEF (each at 1.4 x 10(60 cells) on days 28, 35, and 42 after primary immunization. To test EC-SOD activity, blood samples were collected in each group on day 49. The EC-SOD activity was nearly 1.5-fold higher in the transgenic MEF-treated group than in the nontransgenic MEF-treated group (p < 0.05). The severity of arthritis in mice was scored in a double-blind manner, with each paw being assigned a separate clinical score. The severity of arthritis in EC-SOD transgenic MEF-treated mice was significantly suppressed in the arthritic clinical score (p < 0.05). To investigate the alteration of cytokine levels, ELISA was used to measure blood samples. Levels of IL-1beta and TNF-alpha were reduced in the transgenic MEF-treated group (p < 0.05). Abnormalities of the joints were examined by H&E staining. There were no signs of inflammation except for mild hyperplasia of the synovium in the transgenic MEF-treated group. The proliferation of CII-specific T cells was lower in the transgenic MEF-treated mice than in those in the other groups. The transfer of EC-SOD transgenic MEF has shown a therapeutic effect in CIA mice and this approach may be a safer and more effective form of therapy for rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/surgery , Cell Transplantation/methods , Fibroblasts/transplantation , Superoxide Dismutase/therapeutic use , Animals , Fibroblasts/enzymology , Humans , Keratin-14/genetics , Lymphocyte Activation , Mice , Mice, SCID , Mice, Transgenic , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology
16.
J Interferon Cytokine Res ; 32(1): 6-11, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22175542

ABSTRACT

Calcineurin (CN) is a calcium- and calmodulin-dependent serine/threonine phosphatase. In immune cells, CN controls the activity of a wide range of transcription factors, including nuclear factor of activated T, nuclear factor-kappa B, c-fos, and Elk-1. CN plays an important role in synoviocyte activation and arthritis progression in vivo and this function is tightly linked to dysregulated intracellular Ca(2+) store and Ca(2+) response triggered by proinflammatory cytokines. In the present study, transgenic mice expressing human calcineurin-binding protein 1 (hCabin1) were generated, driven by type II collagen promoter, and the efficiency of these mice was investigated by experimental arthritis. These transgenic mice successfully expressed hCabin1 in joint tissue as well as other organs such as liver, heart, and brain. The overexpression of hCabin1 reduced the disease severity during collagen-induced arthritis. In fibroblast-like synoviocytes (FLSs) from hCabin1 transgenic mice, the productions of these cytokines, including interleukin (IL)-2, IL-4, and IFN-γ, were decreased and matrix metalloproteinases were also depressed in transgenic mice FLS. In addition, these effects were only found in the joint tissue, which is a major inflammation site. These findings will provide a better knowledge of the pathogenic mechanisms of rheumatoid arthritis and a potential animal model of the chronic inflammatory conditions, including atherosclerosis and transplantation.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Synovial Membrane/metabolism , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , Cytokines/biosynthesis , Disease Progression , Gene Expression , Gene Expression Regulation , Humans , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred DBA , Mice, Transgenic , Organ Specificity/genetics
17.
J Invest Dermatol ; 132(4): 1169-76, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22170489

ABSTRACT

Atopic dermatitis (AD) is a chronically relapsing, non contagious pruritic skin disease with two phases: acute and chronic. Cysteine protease cathepsin S (CTSS) is involved in inflammatory processes, possibly leading to atherosclerosis and asthma. Recently, it has been reported that CTSS can arouse a predominant sensation of itch accompanied by classical ligand­receptor signaling [corrected]. Recently, CTSS was shown to be a ligand for proteinase-activated receptor-2 (PAR-2), which is associated with itching. In this study, we show that CTSS-overexpressing transgenic (TG) mice spontaneously develop a skin disorder similar to chronic AD. The results of this study suggest that CTSS overexpression triggers PAR-2 expression in dendritic cells (DCs), resulting in the promotion of CD4(+) differentiation, which is involved in major histocompatibility complex (MHC) class II expression. In addition, we investigated mast cells and macrophages and found significantly higher mean levels of T helper type 1 (Th1) cell-associated cytokines than T helper type 2 (Th2) cell-associated cytokines in CTSS-overexpressing TG mice. These results suggest that increased PAR-2 expression in DCs as a result of CTSS overexpression induces scratching behavior and Th1 cell-associated cytokine expression, and can trigger chronic AD symptoms.


Subject(s)
Cathepsins/metabolism , Dermatitis, Atopic/etiology , Dermatitis, Atopic/metabolism , Disease Models, Animal , Up-Regulation , Animals , CD4-Positive T-Lymphocytes/pathology , Cathepsins/genetics , Cell Proliferation , Chronic Disease , Dendritic Cells/metabolism , Dermatitis, Atopic/pathology , Female , Mast Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptor, PAR-2/genetics , Receptor, PAR-2/metabolism , Signal Transduction/physiology
18.
Exp Mol Med ; 44(9): 529-35, 2012 Sep 30.
Article in English | MEDLINE | ID: mdl-22718219

ABSTRACT

Oxidative stress such as reactive oxygen species (ROS) within the inflamed joint have been indicated as being involved as inflammatory mediators in the induction of arthritis. Correlations between extracellular- superoxide dismutase (EC-SOD) and inflammatory arthritis have been shown in several animal models of RA. However, there is a question whether the over-expression of EC-SOD on arthritic joint also could suppress the progression of disease or not. In the present study, the effect on the synovial tissue of experimental arthritis was investigated using EC-SOD over-expressing transgenic mice. The over-expression of EC- SOD in joint tissue was confirmed by RT-PCR and immunohistochemistry. The degree of the inflammation in EC-SOD transgenic mice was suppressed in the collagen-induced arthritis model. In a cytokine assay, the production of pro-inflammatory cytokines such as, IL-1ß, TNFα, and matrix metalloproteinases (MMPs) was decreased in fibroblast-like synoviocyte (FLS) but not in peripheral blood. Histological examination also showed repressed cartilage destruction and bone in EC-SOD transgenic mice. In conclusion, these data suggest that the over-expression of EC-SOD in FLS contributes to the activation of FLS and protection from joint destruction by depressing the production of the pro-inflammatory cytokines and MMPs. These results provide EC-SOD transgenic mice with a useful animal model for inflammatory arthritis research.


Subject(s)
Arthritis, Experimental/enzymology , Arthritis, Rheumatoid , Superoxide Dismutase , Synovial Fluid/enzymology , Animals , Arthritis, Experimental/blood , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/enzymology , Arthritis, Rheumatoid/pathology , Fibroblasts/metabolism , Gene Expression Regulation , Inflammation/pathology , Interleukin-1beta/blood , Interleukin-1beta/metabolism , Joints/enzymology , Joints/pathology , Matrix Metalloproteinases/blood , Matrix Metalloproteinases/metabolism , Mice , Mice, Transgenic , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Synovial Membrane/pathology
19.
In Vitro Cell Dev Biol Anim ; 46(2): 148-54, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19915933

ABSTRACT

The major objective of this study was to improve the development rate of parthenogenetic porcine embryos. In this study, the anti-oxidative and anti-apoptotic effects of three antioxidants, ß-mercaptoethanol (ß-ME), α-tocopherol, and extracellular superoxide dismutase (EC-SOD), were examined on the development of parthenogenetic porcine embryos. The development rate of parthenogenetic porcine embryos to the blastocyst stage was 8.1% for control; 19.1%, 14.6%, and 5.0% for 1, 3, and 5 µM ß-ME; 17.2% and 17.5% for 50 and 100 µM α-tocopherol and 12.0% and 4.0% for EC-SOD transgenic mouse embryonic fibroblast (Tg-MEF) and EC-SOD non-transgenic mouse embryonic fibroblast (NTg-MEF) conditioned medium at day 3, respectively. Here, ß-ME, α-tocopherol, and EC-SOD Tg-MEF conditioned medium increased the development rate of parthenogenetic porcine embryos to the blastocyst stage (P < 0.05). The average number of total cells and apoptotic cells at the blastocyst was analyzed at the optimal conditions of the three antioxidants. The three antioxidants increased the average number of total cells at the blastocyst, and they decreased apoptotic cells at the blastocyst as compared to control without supplementation (P < 0.05). When the reactive oxygen species levels in two-cell embryos after 1 µM ß-ME and 100 µM α-tocopherol treatment were examined, those were lower than control group (P < 0.05). In conclusion, it was found that the three antioxidants, ß-mercaptoethanol, α-tocopherol, and EC-SOD Tg-MEF, conditioned medium can play a role as a strong stimulator in the development of parthenogenetic porcine embryos.


Subject(s)
Antioxidants/pharmacology , Blastocyst/drug effects , Embryonic Development/drug effects , Swine/embryology , Animals , Apoptosis/drug effects , Culture Media, Conditioned , Embryo Culture Techniques , Mercaptoethanol/pharmacology , Mice , Mice, Transgenic , Parthenogenesis , Reactive Oxygen Species/metabolism , Superoxide Dismutase/pharmacology , alpha-Tocopherol/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL