Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Immunol ; 183(2): 1110-9, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19553542

ABSTRACT

It is not clear why the development of protective Th2 cells is poor in type 1 diabetes (T1D). c-Maf transactivates the IL-4 gene promoting Th2 cell development; therefore, abnormalities in c-Maf may contribute to reduced IL-4 production by CD4 cells from nonobese diabetic (NOD) mice. In this study we demonstrate that despite normal expression, c-Maf binds poorly to the IL-4 promoter (IL-4p) in NOD CD4 cells. Immunoblotting demonstrates that c-Maf can be modified at lysine 33 by SUMO-1 (small ubiquitin-like modifier 1). Sumoylation is facilitated by direct interaction with the E2-conjugating enzyme Ubc9 and increases following T cell stimulation. In transfected cells, sumoylation decreases c-Maf transactivation of IL-4p-driven luciferase reporter activity, reduces c-Maf binding to the IL-4p in chromatin immunoprecipitation assays, and enhances c-Maf localization into promyelocytic leukemia nuclear bodies. Sumoylation of c-Maf is increased in NOD CD4 cells as compared with CD4 cells from diabetes-resistant B10.D2 mice, suggesting that increased c-Maf sumoylation contributes to immune deviation in T1D by reducing c-Maf access to and transactivation of the IL-4 gene.


Subject(s)
Interleukin-4/genetics , Proto-Oncogene Proteins c-maf/physiology , SUMO-1 Protein/immunology , Transcriptional Activation/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Humans , Immunity , Leukemia, Myeloid/pathology , Lysine/metabolism , Mice , Mice, Inbred NOD , Promoter Regions, Genetic , Proto-Oncogene Proteins c-maf/metabolism
2.
Immunology ; 127(2): 267-78, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19476513

ABSTRACT

In addition to transactivation of interleukin-4 (IL-4), cellular muscular aponeurotic fibrosarcoma (c-Maf) enhances CD4 cell apoptosis by limiting Bcl-2 expression. The CD8 cells also express c-Maf and peripheral CD8 cell numbers are reduced in c-Maf transgenic mice, suggesting that c-Maf may influence CD8 cell survival in a manner similar to CD4 cells. Here we confirm that, similar to CD4 cells, c-Maf enhances CD8 cell susceptibility to apoptosis induced by multiple stimuli, independent of IL-4. However, unlike CD4 cells, c-Maf enhancement of apoptosis is independent of Bcl-2, suggesting that c-Maf uses other mechanisms to regulate CD8 cell apoptosis. Real-time reverse transcription-polymerase chain reaction reveals that the pro-apoptotic gene Caspase 6 is upregulated in c-Maf transgenic CD8 cells, suggesting that Caspase 6 is a novel c-Maf target gene. Luciferase reporter assays and site-directed mutagenesis reveal a functional c-Maf recognition element (MARE) within the first intron of Caspase 6. Binding of c-Maf to the MARE site is detectable by chromatin immunoprecipitation using non-transgenic T-cell lysates, so c-Maf can interact with the Caspase 6 MARE site in normal T cells. Furthermore, caspase 6 activity is increased among CD8 cells from c-Maf transgenic mice following T-cell receptor engagement. As expected, activity of the downstream caspases 3 and 7 is also increased. Consistent with the ability of caspase 6 to participate in positive feedback loops, cytochrome c release and caspase 8 activation are also increased. Together these results indicated that c-Maf increases CD8 cell sensitivity to apoptotic stimuli, at least in part, by direct transactivation of Caspase 6, providing increased substrate for Caspase 6-dependent apoptosis pathways.


Subject(s)
Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , Caspase 6/immunology , Proto-Oncogene Proteins c-maf/immunology , Animals , Caspase 6/genetics , Cells, Cultured , Down-Regulation/immunology , Enzyme Activation/immunology , Feedback, Physiological/immunology , Interleukin-4/immunology , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcriptional Activation/immunology
3.
Cell Immunol ; 257(1-2): 88-96, 2009.
Article in English | MEDLINE | ID: mdl-19362713

ABSTRACT

Islet specific CD4 cells expressing inhibitory receptors may be a useful therapeutic tool for treating type 1 diabetes (T1D). Engagement of transgenic Ly49A inhibits CD4 cell activation and delays onset of T1D in mice. However, in vitro studies suggest the inhibitory effect of Ly49A is incomplete. Here we report that following simultaneous TCR and Ly49A engagement, phosphorylation of Zap70, Erk1/2 and c-Jun were significantly diminished. Kinetic studies indicated that Ly49A did not simply delay activation but had a long-lasting effect. In contrast, when only costimulatory signals were provided through CD28, Ly49A engagement did not block p38 MapK or Akt phosphorylation. Likewise, expression of the downstream targets Bcl-xl and Baff were unaffected. Together these data suggest that engagement of Ly49A selectively inhibits signals downstream of the TCR but spares those unique to CD28. These results suggest that when considering its use as an immunotherapy, the potency of inhibitory receptors such as Ly49A may be further improved by pairing them with costimulatory blockade.


Subject(s)
CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/metabolism , NK Cell Lectin-Like Receptor Subfamily A/metabolism , Receptors, Antigen, T-Cell/metabolism , Animals , CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Lymphocyte Activation , Mice , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily A/immunology , Oncogene Protein v-akt/immunology , Oncogene Protein v-akt/metabolism , Phosphorylation/physiology , Proto-Oncogene Proteins c-jun/immunology , Proto-Oncogene Proteins c-jun/metabolism , Receptors, Antigen, T-Cell/immunology , Signal Transduction/physiology , ZAP-70 Protein-Tyrosine Kinase/immunology , ZAP-70 Protein-Tyrosine Kinase/metabolism , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Mol Pain ; 4: 9, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18312687

ABSTRACT

A common complication associated with diabetes is painful or painless diabetic peripheral neuropathy (DPN). The mechanisms and determinants responsible for these peripheral neuropathies are poorly understood. Using both streptozotocin (STZ)-induced and transgene-mediated murine models of type 1 diabetes (T1D), we demonstrate that Transient Receptor Potential Vanilloid 1 (TRPV1) expression varies with the neuropathic phenotype. We have found that both STZ- and transgene-mediated T1D are associated with two distinct phases of thermal pain sensitivity that parallel changes in TRPV1 as determined by paw withdrawal latency (PWL). An early phase of hyperalgesia and a late phase of hypoalgesia are evident. TRPV1-mediated whole cell currents are larger and smaller in dorsal root ganglion (DRG) neurons collected from hyperalgesic and hypoalgesic mice. Resiniferatoxin (RTX) binding, a measure of TRPV1 expression is increased and decreased in DRG and paw skin of hyperalgesic and hypoalgesic mice, respectively. Immunohistochemical labeling of spinal cord lamina I and II, dorsal root ganglion (DRG), and paw skin from hyperalgesic and hypoalgesic mice reveal increased and decreased TRPV1 expression, respectively. A role for TRPV1 in thermal DPN is further suggested by the failure of STZ treatment to influence thermal nociception in TRPV1 deficient mice. These findings demonstrate that altered TRPV1 expression and function contribute to diabetes-induced changes in thermal perception.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Hot Temperature , Pain/metabolism , TRPV Cation Channels/metabolism , Animals , Blood Glucose/metabolism , Body Weight , Diabetes Mellitus, Experimental/physiopathology , Diterpenes/metabolism , Ganglia, Spinal/metabolism , Immunohistochemistry , Injections, Intraperitoneal , Ion Channel Gating , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Pain/physiopathology , Streptozocin , Tritium
5.
Mol Pharmacol ; 73(3): 995-1004, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18089839

ABSTRACT

Streptozotocin (STZ) is a diabetogenic agent extensively used to induce diabetes and to study complications including diabetic peripheral neuropathy (DPN). While studying the influence of transient receptor potential vanilloid 1 (TRPV1) on DPN in the STZ-induced diabetic mouse model, we found that a proportion of STZ-treated mice was nondiabetic but still exhibited hyperalgesia. To understand the mechanism underlying this phenomenon, dorsal root ganglion (DRG) neurons and stably TRPV1 expressing human embryonic kidney (HEK) 293T cells were used to study the expression and function of TRPV1. Incubation of DRG neurons with STZ resulted in a significant increase in the amplitude of capsaicin-induced TRPV1-mediated current and Ca(2+) influx compared with vehicle-treated sister cultures. It was also found that STZ treatment induced higher levels of reactive oxygen species, which was abolished with concomitant treatment with catalase. Treatment of cells with H(2)O(2) mimicked the effects of STZ. Western blot analysis revealed an increase in TRPV1 protein content and phospho p38 (p-p38) mitogen-activated protein kinase (MAPK) levels in DRG of STZ-injected diabetic and nondiabetic hyperalgesic mice compared with control mice. Furthermore, in stably TRPV1-expressing HEK 293T cells, STZ treatment induced an increase in TRPV1 protein content and p-p38 MAPK levels, which was abolished with concomitant treatment with catalase or p38 MAPK inhibitor. These results reveal that STZ has a direct action on neurons and modulates the expression and function of TRPV1, a nociceptive ion channel that is responsible for inflammatory thermal pain.


Subject(s)
Hot Temperature , Hyperalgesia/physiopathology , Neurons, Afferent/drug effects , Streptozocin/pharmacology , TRPV Cation Channels/metabolism , Animals , Calcium/metabolism , Cell Line , Cell Survival/drug effects , Cells, Cultured , Data Interpretation, Statistical , Dose-Response Relationship, Drug , Electrophysiology , Embryo, Mammalian , Female , Ganglia, Spinal/cytology , Humans , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Kidney/cytology , Male , Mice , Neurons, Afferent/metabolism , Pain , Patch-Clamp Techniques , Pregnancy , Rats , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Time Factors
6.
Eur J Immunol ; 37(10): 2868-80, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17823980

ABSTRACT

The transcription factor c-Maf is critical for IL-4 production and the development of Th2 cells, which promote humoral immunity and protect against extracellular parasites. Yet, little else is known of c-Maf function in CD4 cells. Here, we identify a novel role for c-Maf in regulating susceptibility to apoptosis. Overexpression of c-Maf results in increased susceptibility of CD4 cells to apoptosis induced by multiple stimuli, including growth factor withdrawal, dexamethasone, irradiation, and TCR engagement. This effect is independent of Fas or p53; however, Bcl-2 expression is reduced in c-Maf Tg CD4 cells. Immunoprecipitation and Western blot analyses demonstrate that c-Maf-c-Myb complex formation is enhanced among T cells from c-Maf Tg mice compared to non-Tg littermates following TCR engagement. Unlike non-Tg T cells, c-Myb binding to the Bcl-2 promoter is not detectable in c-Maf Tg T cells by chromatin immunoprecipitation. In reporter assays, Bcl-2 promoter activity is reduced by c-Maf in a dose-dependent manner. Furthermore, transgene-mediated Bcl-2 expression corrects the apoptosis defect observed among c-Maf Tg CD4 cells. These data suggest that c-Maf can interact with c-Myb to reduce Bcl-2 expression, thereby limiting CD4 cell survival following TCR engagement.


Subject(s)
Apoptosis/immunology , CD4-Positive T-Lymphocytes/metabolism , Down-Regulation/immunology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-maf/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Animals , Apoptosis/genetics , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Down-Regulation/genetics , Humans , Mice , Mice, Transgenic , Promoter Regions, Genetic/immunology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-maf/physiology , Proto-Oncogene Proteins c-myb/physiology , Receptors, Antigen, T-Cell/metabolism
7.
Nat Med ; 12(8): 933-8, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16862154

ABSTRACT

CD82, also known as KAI1, was recently identified as a prostate cancer metastasis suppressor gene on human chromosome 11p1.2 (ref. 1). The product of CD82 is KAI1, a 40- to 75-kDa tetraspanin cell-surface protein also known as the leukocyte cell-surface marker CD82 (refs. 1,2). Downregulation of KAI1 has been found to be clinically associated with metastatic progression in a variety of cancers, whereas overexpression of CD82 specifically suppresses tumor metastasis in various animal models. To define the mechanism of action of KAI1, we used a yeast two-hybrid screen and identified an endothelial cell-surface protein, DARC (also known as gp-Fy), as an interacting partner of KAI1. Our results indicate that the cancer cells expressing KAI1 attach to vascular endothelial cells through direct interaction between KAI1 and DARC, and that this interaction leads to inhibition of tumor cell proliferation and induction of senescence by modulating the expression of TBX2 and p21. Furthermore, the metastasis-suppression activity of KAI1 was significantly compromised in DARC knockout mice, whereas KAI1 completely abrogated pulmonary metastasis in wild-type and heterozygous littermates. These results provide direct evidence that DARC is essential for the function of CD82 as a suppressor of metastasis.


Subject(s)
Duffy Blood-Group System/metabolism , Endothelium, Vascular/metabolism , Kangai-1 Protein/metabolism , Lung Neoplasms/pathology , Membrane Glycoproteins/metabolism , Neoplasm Metastasis/prevention & control , Receptors, Cell Surface/metabolism , Alleles , Amino Acid Sequence , Animals , Base Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Duffy Blood-Group System/chemistry , Female , Heterozygote , Humans , Kangai-1 Protein/chemistry , Male , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Molecular Sequence Data , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Rats , Receptors, Cell Surface/chemistry , T-Box Domain Proteins/metabolism
8.
J Immunol ; 175(6): 3516-24, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16148094

ABSTRACT

During inflammation, chemokines are conductors of lymphocyte trafficking. The chemokine CXCL10 is expressed early after virus infection. In a virus-induced mouse model for type 1 diabetes, CXCL10 blockade abrogated disease by interfering with trafficking of autoaggressive lymphocytes to the pancreas. We have generated transgenic rat insulin promotor (RIP)-CXCL10 mice expressing CXCL10 in the beta cells of the islets of Langerhans to evaluate how bystander inflammation influences autoimmunity. RIP-CXCL10 mice have islet infiltrations by mononuclear cells and limited impairment of beta cell function, but not spontaneous diabetes. RIP-CXCL10 mice crossed to RIP-nucleoprotein (NP) mice expressing the NP of the lymphocytic choriomeningitis virus in the beta cells had massively accelerated type 1 diabetes after lymphocytic choriomeningitis virus infection. Mechanistically, we found a drastic increase in NP-specific, autoaggressive CD8 T cells in the pancreas after infection. In situ staining with H-2D(b)(NP(396)) tetramers revealed islet infiltration by NP-specific CD8 T cells in RIP-NP-CXCL10 mice early after infection. Our results indicate that CXCL10 expression accelerates the autoimmune process by enhancing the migration of Ag-specific lymphocytes to their target site.


Subject(s)
Chemokines, CXC/genetics , Chemotaxis, Leukocyte , Diabetes Mellitus, Type 1/etiology , Islets of Langerhans/metabolism , Animals , Autoimmunity , CD8-Positive T-Lymphocytes/immunology , Chemokine CXCL10 , Chemokines, CXC/analysis , Chemokines, CXC/physiology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/virology , Inflammation/immunology , Islets of Langerhans/chemistry , Lymphocytic choriomeningitis virus , Mice , Mice, Transgenic , T-Cell Antigen Receptor Specificity , Viral Proteins/immunology
9.
Oncogene ; 24(34): 5389-95, 2005 Aug 11.
Article in English | MEDLINE | ID: mdl-15897909

ABSTRACT

Fatty acid synthase (FAS), a key enzyme of the fatty acid biosynthetic pathway, has been shown to be overexpressed in various types of human cancer and is, therefore, considered to be an attractive target for anticancer therapy. However, the exact mechanism of overexpression of the FAS gene in tumor cells is not well understood. In this report, we demonstrate that the expression of the tumor suppressor gene PTEN has a significant inverse correlation with FAS expression in the case of prostate cancer in the clinical setting, and inhibition of the PTEN gene leads to the overexpression of FAS in vitro. We also found that the combination of the expression status of these two genes is a better prognostic marker than either gene alone. Furthermore, our results indicate that the specific inhibition of FAS gene by siRNA leads to apoptosis of prostate tumor cells, and inhibition of PI 3-kinase pathway synergizes with FAS siRNA to enhance tumor cell death. These results provide a strong rationale for exploring the therapeutic use of an inhibitor of the PTEN signaling pathway in conjunction with the FAS siRNA to inhibit prostate tumor growth.


Subject(s)
Apoptosis , Fatty Acid Synthases/metabolism , Gene Expression Regulation, Neoplastic , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/biosynthesis , Prostatic Neoplasms/genetics , Tumor Suppressor Proteins/biosynthesis , Aged , Aged, 80 and over , Fatty Acid Synthases/biosynthesis , Humans , Immunohistochemistry , Male , Middle Aged , PTEN Phosphohydrolase , Prognosis , Prostatic Neoplasms/pathology , RNA Interference , Signal Transduction , Survival Analysis
10.
Mol Pain ; 1: 17, 2005 Apr 27.
Article in English | MEDLINE | ID: mdl-15857517

ABSTRACT

Insulin and insulin-like growth factors (IGFs) maintain vital neuronal functions. Absolute or functional deficiencies of insulin or IGF-I may contribute to neuronal and vascular complications associated with diabetes. Vanilloid receptor 1 (also called TRPV1) is an ion channel that mediates inflammatory thermal nociception and is present on sensory neurons. Here we demonstrate that both insulin and IGF-I enhance TRPV1-mediated membrane currents in heterologous expression systems and cultured dorsal root ganglion neurons. Enhancement of membrane current results from both increased sensitivity of the receptor and translocation of TRPV1 from cytosol to plasma membrane. Receptor tyrosine kinases trigger a signaling cascade leading to activation of phosphatidylinositol 3-kinase (PI(3)K) and protein kinase C (PKC)-mediated phosphorylation of TRPV1, which is found to be essential for the potentiation. These findings establish a link between the insulin family of trophic factors and vanilloid receptors.


Subject(s)
Insulin-Like Growth Factor I/physiology , Insulin/physiology , TRPV Cation Channels/metabolism , Animals , Cell Line , Humans , Protein Transport/physiology , Rats , Signal Transduction/physiology , TRPV Cation Channels/physiology , Xenopus laevis
11.
J Immunol ; 174(7): 3897-905, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15778344

ABSTRACT

Activation of islet-specific T cells plays a significant role in the development of type 1 diabetes. In an effort to control T cell activation, we expressed the inhibitory receptor, Ly-49A, on islet-specific mouse CD4 cells. Ag-mediated activation of Ly-49A T cells was inhibited in vitro when the Ly-49A ligand, H-2D(d), was present on APCs. Ag-driven T cell proliferation, cytokine production, and changes in surface receptor expression were significantly reduced. Inhibition was also evident during secondary antigenic challenge. Addition of exogenous IL-2 did not rescue cells from inhibition, suggesting that Ly-49A engagement does not lead to T cell anergy. Importantly, in an adoptive transfer model, Ly-49A significantly delays the onset of diabetes. Together these results demonstrate that the inhibitory receptor Ly-49A effectively limits Ag-specific CD4 cell responses even in the presence of sustained autoantigen expression in vivo.


Subject(s)
Antigens, Ly/pharmacology , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/prevention & control , Lymphocyte Activation/drug effects , Adoptive Transfer , Animals , Antigen Presentation , Antigens, Ly/genetics , Autoantigens/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/transplantation , Cytokines/biosynthesis , Cytokines/drug effects , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , H-2 Antigens , Histocompatibility Antigen H-2D , Interleukin-2/pharmacology , Islets of Langerhans/immunology , Lectins, C-Type , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Receptors, NK Cell Lectin-Like
12.
Diabetes ; 53(4): 978-88, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15047613

ABSTRACT

We have produced a T-cell receptor (TCR) transgenic NOD mouse, 6.9TCR/NOD, in which the expression of both diabetogenic T-cells and naturally occurring autoantigen were simultaneously controlled. The parent T-cell clone, BDC-6.9, and T-cells from 6.9TCR/NOD mice recognize a currently unidentified antigen present in NOD but not in BALB/c islet cells. A gene that codes for the antigen, or a protein that regulates the antigen, was previously mapped to a locus on chromosome 6. We have developed transgenic mice bearing the TCR alpha- and beta-chains from the BDC-6.9 T-cell clone on a NOD congenic background in which the antigen locus on chromosome 6 of the NOD mouse is replaced by a segment from BALB/c. These NOD.C6 congenic mice lack the NOD islet cell antigen to which the BDC-6.9 T-cell clone responds. Diabetes in both male and female 6.9TCR/NOD mice is dramatically accelerated, but in 6.9TCR/NOD.C6 mice lacking the NOD islet cell autoantigen, we have not observed diabetes for up to 1 year of age. Thus, the generation of 6.9TCR transgenic mice provides a model of autoimmune diabetes whereby controlled expression of an endogenous polymorphic autoantigen effectively determines disease development.


Subject(s)
Autoantigens/genetics , Diabetes Mellitus/genetics , Polymorphism, Genetic , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chromosome Mapping , DNA Primers , Diabetes Mellitus/immunology , Disease Susceptibility , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor/genetics , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Transgenic , Promoter Regions, Genetic , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...