Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 152
Filter
1.
Genes Immun ; 11(3): 219-31, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20220769

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic debilitating disease resulting from a complex interaction of multiple genetic factors with the environment. To identify modifier genes of IBD, we used an F2 intercross of IBD-resistant C57BL/6J-Il10(-/-) mice and IBD-susceptible C3H/HeJBir-Il10(-/-) (C3Bir-Il10(-/-)) mice. We found a prominent involvement of lymphatic vessels in IBD and applied a scoring system to quantify lymphatic vascular changes. Quantitative trait locus (QTL) analyses revealed a large-effect QTL on chromosome 3, mapping to an interval of 43.6 Mbp. This candidate interval was narrowed by fine mapping to 22 Mbp, and candidate genes were analyzed by a systems genetics approach that included quantitative gene expression profiling, search for functional polymorphisms, and haplotype block analysis. We identified vascular adhesion molecule 1 (Vcam1) as a candidate modifier gene in the interleukin 10-deficient mouse model of IBD. Importantly, VCAM1 protein levels were increased in susceptible C3H/HeJ mice, compared with C57BL/6J mice; systemic blockade of VCAM1 in C3Bir-Il10(-/-) mice reduced their inflammatory lymphatic vessel changes. These results indicate that genetically determined expression differences of VCAM1 are associated with susceptibility to colon inflammation, which is accompanied by extensive lymphatic vessel changes. VCAM1 is, therefore, a promising therapeutic target for IBD.


Subject(s)
Genetic Predisposition to Disease , Inflammatory Bowel Diseases/genetics , Quantitative Trait Loci/genetics , Vascular Cell Adhesion Molecule-1/genetics , Animals , Chromosome Mapping , Chromosomes, Mammalian/genetics , Female , Gene Expression Profiling , Haplotypes , Inflammatory Bowel Diseases/metabolism , Interleukin-10/deficiency , Interleukin-10/genetics , Lod Score , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Polymorphism, Single Nucleotide , Reverse Transcriptase Polymerase Chain Reaction
2.
Diabetes Obes Metab ; 9 Suppl 2: 14-22, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17919174

ABSTRACT

We describe unexpected alterations in the non-obese diabetic (NOD/Lt) mouse model of type 1 diabetes (T1D) following forced beta-cell expression of non-mammalian genes ligated to an insulin promoter sequence. These include the jellyfish green fluorescent protein (GFP), useful for beta-cell identification, and the bacteriophage P1 Cre recombinase, necessary for beta cell-specific ablation of a gene using a Cre-loxP system. Homozygous expression of GFP, driven by the mouse insulin 1 gene promoter (MIP-GFP) in a single transgenic line of NOD mice, produced T1D in postnatal mice that was not associated with insulitis, but rather beta cell-depleted islets. Hemizygous transgene expression suppressed spontaneous autoimmune T1D in females, and produced a male glucose intolerance syndrome associated with age-dependent declines in plasma insulin content. Among lines of transgenic NOD/Lt mice expressing Cre recombinase driven by the rat insulin 2 promoter (RIP-Cre), high, non-mosaic expression correlated with suppressed T1D development. These findings emphasize the need for careful characterization of genetically manipulated NOD mouse stocks to insure that model characteristics have not been compromised.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 1/genetics , Insulin-Secreting Cells/immunology , Mice, Transgenic/immunology , Animals , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Female , Gene Expression , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Integrases/genetics , Integrases/metabolism , Male , Mice , Mice, Inbred NOD , Mice, Transgenic/genetics , Pancreas/pathology , Transgenes
3.
Tissue Antigens ; 70(2): 87-95, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17610413

ABSTRACT

It has recently become apparent that several molecules involved in the control of metabolism also play an important function in the regulation of immune responses. Among those molecules, the adipocyte-derived cytokine leptin has been shown to significantly influence innate and adaptive immune responses both in normal and in pathological conditions. For example, levels of leptin are typically low in infection and high in autoimmunity, both systemically and at the site of inflammation. Moreover, in addition to its long-known effects on the promotion of T helper 1 immune responses and cell-mediated immunity, leptin has more recently been found capable to constrain proliferation of regulatory T cells. As such, leptin represents not only a link between metabolism and immune responses in general but also a pivotal modulator of the magnitude of selected mechanisms of peripheral immunity in relation to body fat mass. We review here the most recent advances on the role of leptin in the control of immune tolerance and critically discuss how strategies aimed at neutralizing the leptin axis could represent innovative tools for the therapy of autoimmune disorders.


Subject(s)
Adipose Tissue/immunology , Adipose Tissue/metabolism , Autoimmune Diseases/metabolism , Leptin/physiology , Animals , Humans
4.
Genes Immun ; 6(6): 528-38, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16015371

ABSTRACT

While sharing the H2g7 MHC and many other important Type I diabetes susceptibility (Idd) genes with NOD mice, the NOR strain remains disease free due to resistance alleles within the approximately 12% portion of their genome that is of C57BLKS/J origin. Previous F2 segregation analyses indicated multiple genes within the 'Idd13' locus on Chromosome 2 provide the primary component of NOR diabetes resistance. However, it was clear other genes also contribute to NOR diabetes resistance, but were difficult to detect in the original segregation analyses because they were relatively weak compared to the strong Idd13 protection component. To identify these further genetic components of diabetes resistance, we performed a new F2 segregation analyses in which NOD mice were outcrossed to a 'genome-conditioned' NOR stock in which a large component of Idd13-mediated resistance was replaced with NOD alleles. These F2 segregation studies combined with subsequent congenic analyses confirmed the presence of additional NOR resistance genes on Chr. 1 and Chr. 4, and also potentially on Chr. 11. These findings emphasize the value for diabetes gene discovery of stratifying not only MHC loci conferring the highest relative risk but also as many as possible of the non-MHC loci presumed to contribute significantly.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease , Genome , Alleles , Animals , Chromosome Mapping , Female , Mice , Mice, Congenic , Mice, Inbred NOD
5.
Diabetologia ; 48(2): 261-7, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15692809

ABSTRACT

AIMS/HYPOTHESIS: ALR/Lt, a mouse strain with strong resistance to type 1 diabetes, is closely related to autoimmune type 1 diabetes-prone NOD/Lt mice. ALR pancreatic beta cells are resistant to the beta cell toxin alloxan, combinations of cytotoxic cytokines, and diabetogenic NOD T-cell lines. Reciprocal F1 hybrids between either ALR and NOD or ALR and NON/Lt, showed that alloxan resistance was transmitted to F1 progeny only when ALR was the maternal parent. Here we show that the mitochondrial genome (mtDNA) of ALR mice contributes resistance to diabetes. METHODS: When F1 progeny from reciprocal outcrosses between ALR and NOD were backcrossed to NOD, a four-fold lower frequency of spontaneous type 1 diabetes development occurred when ALR contributed the mtDNA. Because of the apparent interaction between nuclear and mtDNA, the mitochondrial genomes were sequenced. RESULTS: An ALR-specific sequence variation in the mt-Nd2 gene producing a leucine to methionine substitution at amino acid residue 276 in the NADH dehydrogenase 2 was discovered. An isoleucine to valine mutation in the mt-Co3 gene encoding COX3 distinguished ALR and NOD from NON and ALS. All four strains were distinguished by variation in a mt-encoded arginyl tRNA polyadenine tract. Shared alleles of mt-Co3 and mt-Tr comparing NOD and ALR allowed for exclusion of these two genes as candidates, implicating the mt-Nd2 variation as a potential ALR-derived type 1 diabetes protective gene. CONCLUSIONS/INTERPRETATION: The unusual resistance of ALR mice to both ROS-mediated and autoimmune type 1 diabete stresses reflects an interaction between the nuclear and mt genomes. The latter contribution is most likely via a single nucleotide polymorphism in mt-Nd2.


Subject(s)
DNA, Mitochondrial/genetics , Diabetes Mellitus, Type 1/genetics , NADH Dehydrogenase/genetics , Animals , Base Sequence , Crosses, Genetic , DNA Primers , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Female , Genetic Variation , Immunity, Innate , Kidney/enzymology , Male , Mice , Mice, Inbred NOD , Mice, Mutant Strains , Mitochondria/enzymology , Mitochondria/genetics , Mitochondria, Liver/enzymology , Mitochondria, Liver/genetics , Polymerase Chain Reaction , Polymorphism, Single Nucleotide
7.
Diabetologia ; 45(3): 296-308, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11914735

ABSTRACT

The mouse has been a favoured organism for molecular manipulation in studies seeking to establish the genetic bases and pathophysiologic mechanisms underlying both Type I (insulin-dependent) and Type II (non-insulin-dependent) diabetes mellitus. Gene targeting and transgenesis are the two powerful molecular technologies used in these endeavours. Interpretation of results generated from such studies, either entailing an altered phenotype or the absence of a phenotypic change, is not always simple. This review focuses on certain complications inherent in the methodologies, and outlines steps that can be taken to distinguish effects of the genetic manipulation from unexpected contributions from the genetic background.


Subject(s)
Diabetes Mellitus/genetics , Mice, Knockout/genetics , Mice, Transgenic/genetics , Animals , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Glucose/metabolism , Mice
8.
Proc Natl Acad Sci U S A ; 98(24): 13820-5, 2001 Nov 20.
Article in English | MEDLINE | ID: mdl-11707574

ABSTRACT

Colitic lesions are much more severe in C3H/HeJBir (C3H) than C57BL/6J (B6) mice after 10 backcrosses of a disrupted interleukin-10 (Il10) gene. This study identified cytokine deficiency-induced colitis susceptibility (Cdcs) modifiers by using quantitative trait locus (QTL) analysis. A segregating F(2) population (n = 408) of IL-10-deficient mice was genotyped and necropsied at 6 weeks of age. A major C3H-derived colitogenic QTL (Cdcs1) on chromosome (Chr.) 3 contributed to lesions in both cecum [logarithm of odds ratio (LOD) = 14.6)] and colon (LOD = 26.5) as well as colitis-related phenotypes such as spleen/body weight ratio, mesenteric lymph node/body weight ratio, and secretory IgA levels. Evidence for other C3H QTL on Chr. 1 (Cdcs2) and Chr. 2 (Cdcs3) was obtained. Cdcs1 interacted epistatically or contributed additively with loci on other chromosomes. The resistant B6 background also contributed colitogenic QTL: Cdcs4 (Chr. 8), Cdcs5 (Chr. 17, MHC), and Cdcs6 (Chr. 18). Epistatic interactions between B6 QTL on Chr. 8 and 18 contributing to cecum hyperplasia were particularly striking. In conclusion, a colitogenic susceptibility QTL on Chr. 3 has been shown to exacerbate colitis in combination with modifiers contributed from both parental genomes. The complex nature of interactions among loci in this mouse model system, coupled with separate deleterious contributions from both parental strains, illustrates why detection of human inflammatory bowel disease linkages has proven to be so difficult. A human ortholog of the Chr. 3 QTL, if one exists, would map to Chr. 4q or 1p.


Subject(s)
Colitis/genetics , Interleukin-10/immunology , Quantitative Trait, Heritable , Animals , Chromosomes , Colitis/pathology , Colitis/physiopathology , Epistasis, Genetic , Female , Genetic Linkage , Interleukin-10/genetics , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout
9.
Gene ; 275(2): 267-77, 2001 Sep 19.
Article in English | MEDLINE | ID: mdl-11587854

ABSTRACT

Mono(ADP-ribosyl)transferases regulate the function of target proteins by attaching ADP-ribose to specific amino acid residues in their target proteins. The purpose of this study was to determine the structure, chromosomal localization, and expression profile of the gene for mouse ecto-ADP-ribosyltransferase ART5. Southern blot analyses indicate that Art5 is a single copy gene which maps to mouse chromosome 7 at offset 49.6 cM in close proximity to the Art1, Art2a and Art2b genes. Northern blot and RT-PCR analyses demonstrate prominent expression of Art5 in testis, and lower levels in cardiac and skeletal muscle. Sequence analyses reveal that the Art5 gene encompasses six exons spanning 8 kb of genomic DNA. The 5' end of the Art5 gene overlaps with that of the Art1 gene. A single long exon encodes the predicted ART5 catalytic domain. Separate exons encode the N-terminal leader peptide and a hydrophilic C-terminal extension. Sequencing of RT-PCR products and ESTs identified six splice variants. The deduced amino acid sequence of ART5 shows 87% sequence identity to its orthologue from the human, and 37 and 32% identity to its murine paralogues ART1 and ART2. Unlike ART1 and ART2, ART5 lacks a glycosylphosphatidylinositol-anchor signal sequence and is predicted to be a secretory enzyme. This prediction was confirmed by transfecting an Art5 cDNA expression construct into Sf9 insect cells. The secreted epitope-tagged ART5 protein resembled rat ART2 in exhibiting potent NAD-glycohydrolase activity. This study provides important experimental tools to further elucidate the function of ART5.


Subject(s)
ADP Ribose Transferases/genetics , Genes/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Cell Line , Chromosome Mapping , DNA/chemistry , DNA/genetics , DNA, Complementary/chemistry , DNA, Complementary/genetics , Exons , Female , Gene Expression , Introns , Isoenzymes/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Molecular Sequence Data , Muridae , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Tissue Distribution
11.
J Immunol ; 167(4): 2404-10, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11490031

ABSTRACT

Transgenic restoration of normally absent H2-E MHC class II molecules on APC dominantly inhibits T cell-mediated autoimmune diabetes (IDDM) in nonobese diabetic (NOD) mice. We analyzed the minimal requirements for transgenic H2-E expression on APC subtypes (B lymphocytes vs macrophages/dendritic cells (DC)) to inhibit IDDM. This issue was addressed through the use of NOD stocks transgenically expressing high levels of H2-E and/or made genetically deficient in B lymphocytes in a series of genetic intercross and bone marrow/lymphocyte chimera experiments. Standard (H2-E(null)) NOD B lymphocytes exert a pathogenic function(s) necessary for IDDM. However, IDDM was inhibited in mixed chimeras where H2-E was solely expressed on all B lymphocytes. Interestingly, this resistance was abrogated when even a minority of standard NOD H2-E(null) B lymphocytes were also present. In contrast, in NOD chimeras where H2-E expression was solely limited to approximately half the macrophages/DC, an active immunoregulatory process was induced that inhibited IDDM. Introduction of a disrupted IL-4 gene into the NOD-H2-E transgenic stock demonstrated that induction of this Th2 cytokine does not represent the IDDM protective immunoregulatory process mediated by H2-E expression. In conclusion, high numbers of multiple subtypes of APC must express H2-E MHC class II molecules to additively inhibit IDDM in NOD mice. This raises a high threshold for success in future intervention protocols designed to inhibit IDDM by introduction of putatively protective MHC molecules into hemopoietic precursors of APC.


Subject(s)
Antigen-Presenting Cells/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/prevention & control , HLA-DR Antigens/genetics , Transgenes/immunology , Animals , Antigen-Presenting Cells/classification , Antigen-Presenting Cells/metabolism , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Diabetes Mellitus, Type 1/immunology , Female , Gene Expression Regulation/immunology , H-2 Antigens/biosynthesis , H-2 Antigens/genetics , HLA-DR Antigens/biosynthesis , Immunity, Innate/genetics , Interleukin-4/biosynthesis , Interleukin-4/physiology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred NOD , Mice, Transgenic , Sequence Homology, Amino Acid , Th2 Cells/immunology , Th2 Cells/metabolism
12.
Proc Natl Acad Sci U S A ; 98(1): 235-40, 2001 Jan 02.
Article in English | MEDLINE | ID: mdl-11136257

ABSTRACT

Genetic analysis of autoimmune insulin-dependent diabetes mellitus (IDDM) has focused on genes controlling immune functions, with little investigation of innate susceptibility determinants expressed at the level of target beta cells. The Alloxan (AL) Resistant (R) Leiter (Lt) mouse strain, closely related to the IDDM-prone nonobese diabetic (NOD)/Lt strain, demonstrates the importance of such determinants. ALR mice are unusual in their high constitutive expression of molecules associated with dissipation of free-radical stress systemically and at the beta-cell level. ALR islets were found to be remarkably resistant to two different combinations of beta-cytotoxic cytokines (IL-1beta, tumor necrosis factor alpha, and IFN-gamma) that destroyed islets from the related NOD and alloxan-susceptible strains. The close MHC relatedness between the NOD and ALR strains (H2-Kd and H2-Ag7 identical) allowed us to examine whether ALR islet cells could survive autoimmune destruction by NOD-derived Kd-restricted diabetogenic cytotoxic T lymphocyte clones (AI4 and the insulin-reactive G9C8 clones). Both clones killed islet cells from all Kd-expressing strains except ALR. ALR resistance to diabetogenic immune systems was determined in vivo by means of adoptive transfer of the G9C8 clone or by chimerizing lethally irradiated ALR or reciprocal (ALR x NOD)F1 recipients with NOD bone marrow. In all in vivo systems, ALR and F1 female recipients of NOD marrow remained IDDM free; in contrast, all of the NOD recipients became diabetic. In conclusion, the ALR mouse presents a unique opportunity to identify dominant IDDM resistance determinants expressed at the beta cell level.


Subject(s)
Autoimmunity/immunology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Genetic Predisposition to Disease , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Adoptive Transfer , Alloxan/pharmacology , Animals , Autoimmunity/genetics , Bone Marrow Transplantation/immunology , Cell Death/drug effects , Chimera/genetics , Chimera/immunology , Clone Cells/immunology , Clone Cells/metabolism , Cyclophosphamide/pharmacology , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/chemically induced , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Female , Histocompatibility Antigens Class I/analysis , Histocompatibility Antigens Class I/immunology , Insulin/metabolism , Insulin Secretion , Interferon-gamma/biosynthesis , Interferon-gamma/pharmacology , Interferon-gamma/toxicity , Interleukin-1/pharmacology , Interleukin-1/toxicity , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Lymphocyte Activation , Mice , Mice, Inbred NOD , Mice, Inbred Strains , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/toxicity
13.
Genome Res ; 10(10): 1568-78, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11042154

ABSTRACT

Obesity, a major risk factor for type II diabetes, is becoming more prevalent in Western populations consuming high calorie diets while expending less energy both at the workplace and at home. Most human obesity, and probably most type II diabetes as well, reflects polygenic rather than monogenic inheritance. We have genetically dissected a polygenic mouse model of obesity-driven type II diabetes by outcrossing the obese, diabetes-prone, NZO (New Zealand Obese)/HlLt strain to the relatively lean NON (Nonobese Nondiabetic)/Lt strain, and then reciprocally backcrossing obese F1 mice to the lean NON/Lt parental strain. A continuous distribution of body weights was observed in a population of 203 first backcross males. The 22% of first backcross males developing overt diabetes showed highest peripubertal weight gains and earliest development of hyperinsulinemia. We report a complex diabetes-predisposing ("diabesity") QTL (Quantitative Trait Loci) on chromosome 1 contributing significant main effects to increases in body weight, plasma insulin, and plasma glucose. NZO contributed QTL with significant main effects on adiposity parameters on chromosomes 12 and 5. A NON QTL on chromosome 14 interacted epistatically with the NZO obesity QTL on chromosome 12 to increase adiposity. Although the main effect of the diabetogenic QTL on chromosome 1 was on rapid growth rather than adiposity, it interacted epistatically with the obesity QTL on chromosome 12 to increase plasma glucose levels. Additional complex epistatic interactions eliciting significant increases in body weight and/or plasma glucose were found between the NZO-contributed QTL on chromosome 1 and other NZO-contributed QTL on chromosomes 15 and 17, as well as with an NON-contributed QTL on chromosome 2. We further show that certain of these intergenic interactions are predicated on, or enhanced by, the maternal postparturitional environment. We show by cross-fostering experiments that the maternal environmental influence in part is because of the presence of early obesity-inducing factors in the milk of obese F1 dams. We also discuss a strategy for using recombinant congenic strains to separate and reassemble interacting QTL for future study.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Maternal-Fetal Exchange/genetics , Animals , Chromosome Segregation/genetics , Crosses, Genetic , Female , Genes, Dominant/genetics , Genetic Markers/genetics , Genotype , Lactation/genetics , Male , Mice , Mice, Inbred NOD , Mice, Obese , Penetrance , Pregnancy , Quantitative Trait, Heritable
14.
J Immunol ; 165(8): 4463-9, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11035085

ABSTRACT

T cells proteolytically shed the ectodomains of several cell surface proteins and, thereby, can alter their responsiveness and can release soluble intercellular regulators. ART2.2 is a GPI-anchored ecto-ADP-ribosyltransferase (ART) related to ADP-ribosylating bacterial toxins. ART2.2 is expressed exclusively by mature T cells. Here we show that ART2.2 is shed from the cell surface in enzymatically active form upon activation of T cells. Shedding of ART2.2 resembles that of L-selectin (CD62L) in dose response, kinetics of release, and sensitivity to the metalloprotease inhibitor Immunex Compound 3, suggesting that ART2.2, like CD62L, is cleaved by TNF-alpha-converting enzyme or by another metalloprotease. ART2.2 shed from activated T cells migrates slightly faster in SDS-PAGE analyses than does ART2.2 released upon cleavage of the GPI anchor. This indicates that shedding of ART2.2 is mediated by proteolytic cleavage close to its membrane anchor. Shed ART2.2 is enzymatically active and ADP-ribosylates several substrates in vitro. Thus, shedding of ART2.2 releases a potential intercellular regulator. Finally, using a new FACS assay for monitoring ADP-ribosylation of cell surface proteins, we demonstrate that shedding of ART2.2 correlates with a reduced sensitivity of T cell surface proteins to ADP-ribosylation. Our findings suggest that by shedding ART2.2 the activated T cell not only releases a potential intercellular regulator but also may alter its responsiveness to immune regulation by ART2.2-mediated ADP-ribosylation of cell surface proteins.


Subject(s)
ADP Ribose Transferases , Histocompatibility Antigens/metabolism , Lymphocyte Activation , Membrane Glycoproteins , Metalloendopeptidases/physiology , Poly(ADP-ribose) Polymerases/metabolism , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Adenosine Diphosphate Ribose/metabolism , Animals , Antigens, Differentiation, T-Lymphocyte , Cell Membrane/enzymology , Cell-Free System/immunology , Dose-Response Relationship, Immunologic , Enzyme Activation/immunology , Enzyme Inhibitors/pharmacology , Glycosylphosphatidylinositols/metabolism , Hydrolysis , Kinetics , L-Selectin/metabolism , Lymphocyte Activation/drug effects , Membrane Proteins/metabolism , Metalloendopeptidases/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Molecular Weight , Phosphatidylinositol Diacylglycerol-Lyase , Precipitin Tests , Protein Kinase C/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/enzymology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured , Type C Phospholipases/metabolism
15.
Diabetes ; 49(1): 131-4, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10615961

ABSTRACT

The common Kd and/or Db alleles of NOD mice contribute to the development of autoimmune diabetes, but their respective contributions are unresolved. The major histocompatibility complex (MHC) of the CTS/Shi mouse, originally designated as H2ct, shares MHC class II region identity with the H2g7 haplotype of NOD mice. However, CTS mice were reported to express distinct but undefined MHC class I gene products. Because diabetes frequency was reduced 56% in females of a NOD stock congenic for H2ct, this partial resistance may have derived from the MHC class I allelic differences. In the present report, we use a combination of serologic analysis and sequencing of MHC class I cDNAs to establish that NOD/Lt and CTS/Shi share a common H2-Kd allele but differ at the H2-D end of the MHC complex. The H2-D allele of CTS/Shi was identified as the rare H2-Ddx recently described in ALR/Lt, another NOD-related strain. These results in mouse model systems show that multiple MHC genes confer diabetes resistance and suggest that at least one of the protective MHC or MHC-linked genes in CTS mice may be at the H2-D end of the complex.


Subject(s)
Major Histocompatibility Complex/genetics , Mice, Inbred NOD/genetics , Mice, Inbred Strains/genetics , Alleles , Animals , Diabetes Mellitus/genetics , Flow Cytometry , Haplotypes/genetics , Histocompatibility Antigens Class II/analysis , Immunity, Innate/genetics , Leukocytes/immunology , Mice , Microsatellite Repeats/genetics , Polymorphism, Genetic/genetics , Spleen/cytology , Spleen/immunology
16.
Inflamm Bowel Dis ; 6(4): 290-302, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11149562

ABSTRACT

Severity of inflammatory bowel disease in IL-10 gene-targeted mice is in part determined by genetic background. In the current study, a targeted IL-10 gene was transferred into the C3H/HeJBir substrain, known to exhibit high T-cell and B-cell responses to enteric flora, and to be highly sensitive to colitigenic stress. IL-10-deficient C3H/HeJBir mice developed early onset colitis in contrast to IL-10-deficient C57BL/6J congenic mice. Histopathologic analysis of disease in C3H/HeJBir.Il10-/- and C57BL/6J.Il10-/- mice showed significant differences at all ages studied. Hybrids of these congenic strains (F1.Il10-/-) were produced to study the mode of inheritance as well as subphenotypes that correlated with histopathology. Lesions in F1 mice were intermediate between parental strains. C3H-contributed subphenotypes that correlated best with histopathology were peripheral blood granulocyte percentage, serum amyloid A concentration, spleen weight/body weight ratio, and mesenteric lymph node weight/ body weight ratio. Neither enhanced humoral immunity (secretory IgA, anti-Escherichia coli cellular membrane Ig) characteristic of C3H/HeJBir, nor T-cell percentages in peripheral blood correlated as well. This study represents a necessary step in elucidating murine genetic modifiers controlling colitis sensitivity.


Subject(s)
Colitis, Ulcerative/genetics , Colitis, Ulcerative/pathology , Interleukin-10/deficiency , Interleukin-10/genetics , Animals , Antigen-Presenting Cells , CD4-Positive T-Lymphocytes , Colitis, Ulcerative/immunology , Cytokines , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Genetic Predisposition to Disease , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL
17.
Immunogenetics ; 51(3): 206-11, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10752630

ABSTRACT

The mouse Fcgr1 gene encoding the high-affinity IgG receptor (FcgammaRI) exists as two known alleles, FcgammaRI-BALB and FcgammaRI-NOD, and these alleles exhibit functional differences. To determine whether other alleles exist in mouse strains, Fcgr1 coding regions from 35 strains of mice were sequenced and a further five alleles were identified. The FcgammaRI-BALB and NOD alleles are now designated the "a" and "d" alleles, respectively. Analysis of the five new alleles revealed that although no polymorphisms were observed in the two leader exons, nucleotide and subsequent amino acid changes were observed in the exons encoding the extracellular domains, and transmembrane and cytoplasmic tail. The cDNA of the seven alleles (a-g) were isolated and transiently transfected into COS cells, and IgG-binding studies were performed. Receptors encoded by four of the five new alleles (b, c, f, g) bound IgG2a with high affinity, displaying IgG binding characteristics similar to the a allele (previously FcgammaRI-BALB). The d allele (previously FcgammaRI-NOD) and the e allele [derived from Mus spretus (SPRET/Ei)] encoded receptors which showed broader specificity by binding monomeric IgG2a, IgG2b, and IgG3.


Subject(s)
Alleles , Receptors, IgG/genetics , Amino Acid Sequence , Animals , Base Sequence , COS Cells , DNA, Complementary , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Inbred Strains , Molecular Sequence Data , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
18.
J Immunol ; 163(11): 6014-22, 1999 Dec 01.
Article in English | MEDLINE | ID: mdl-10570289

ABSTRACT

ADP-ribosylation of membrane proteins on mouse T cells by ecto-ADP-ribosyltransferase(s) (ARTs) can down-regulate proliferation and function. The lack of mAbs against mouse ARTs has heretofore prevented analysis of ART expression on T cell subsets. Using gene gun technology, we immunized a Wistar rat with an Art2b expression vector and produced a novel mAb, Nika102, specific for ART2.2, the Art2b gene product. We show that ART2.2 is expressed as a GPI-anchored protein on the surface of mature T cells. Inbred strain-dependent differences in ART2.2 expression levels were observed. C57BL/6J and C57BLKS/J express the Ag at high level, with up to 70% of CD4+ and up to 95% of CD8+ peripheral T cells expressing ART2.2. CBA/J and DBA/2J represent strains with lowest expression levels. T cell-deficient mice and NZW/LacJ mice with a defective structural gene for this enzyme were ART2.2 negative. In the thymus, ART2.2 expression is restricted to subpopulations of mature cells. During postnatal ontogeny, increasing percentages of T cells express ART2.2, reaching a peak at 6-8 wk of age. Interestingly, ART2.2 and CD25 are reciprocally expressed: activation-induced up-regulation of CD25 is accompanied by loss of ART2.2 from the cell surface. Nika102 thus defines a new differentiation/activation marker of thymic and postthymic T cells in the mouse and should be useful for further elucidating the function of the ART2.2 cell surface enzyme.


Subject(s)
ADP Ribose Transferases , Antigens, Differentiation, T-Lymphocyte/immunology , Histocompatibility Antigens/immunology , Membrane Glycoproteins , Mice, Inbred Strains/immunology , Poly(ADP-ribose) Polymerases/immunology , T-Lymphocyte Subsets/enzymology , Animals , Antibodies, Monoclonal , Antibody Specificity , Antigens, Differentiation, T-Lymphocyte/genetics , Down-Regulation , Histocompatibility Antigens/genetics , Lymphocyte Activation , Membrane Proteins/immunology , Mice , Oligopeptides , Peptides , Phosphatidylinositol Diacylglycerol-Lyase , Poly(ADP-ribose) Polymerases/genetics , Rats , Recombinant Proteins/immunology , Thymus Gland/cytology , Thymus Gland/growth & development , Type C Phospholipases/metabolism
19.
Diabetes ; 48(11): 2189-96, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10535453

ABSTRACT

ALS/Lt and ALR/Lt are inbred mouse strains selected for susceptibility and resistance to alloxan (AL)-induced diabetes. Within 24-h after AL administration in vivo, ALS/Lt islets were distinguished from ALR/Lt islets by more extensive necrotic changes. Within 7 days post-AL, ALS/Lt mice exhibited hyperglycemia and hypoinsulinemia, whereas ALR/Lt mice maintained normal plasma insulin and glucose levels. We have recently shown that resistance in ALR/Lt correlated with constitutively elevated systemic (and pancreatic) free radical defense status. In the present report, we examined whether ability to detoxify free radical stress extended to the level of ALR/Lt pancreatic islets. Cultured ALS/Lt islets exposed for 5 min to increasing (0-3 mmol/l) AL concentrations in vitro exhibited an 80% decline in numbers of intact islets after a subsequent 6-day culture period, as well as a 75% reduction in islet insulin content and a 94% decrease in glucose-stimulated insulin secretory capacity. In contrast, ALR/Lt islets remained viable and retained glucose-stimulated insulin secretory capacity as well as normal insulin content. This ALR/Lt islet resistance extended to hydrogen peroxide, a free radical generator whose entry into beta-cells is not dependent on glucose transporters. The elevated antioxidant defenses previously found in ALR/Lt pancreas were extended to isolated islets, which exhibited significantly higher glutathione and Cu-Zn superoxide dismutase 1 levels compared with ALS/Lt islets. A dominant genetic trait from ALR/Lt controlling this unusual AL resistance was indicated by the finding that reciprocal F1 mice of both sexes were resistant to AL administration in vivo. A backcross to ALS/Lt showed 1:1 segregation for susceptibility/resistance, indicative of a single gene controlling the phenotype. In conclusion, the ALR/Lt mouse may provide important insight into genetic mechanisms capable of rendering islets strongly resistant to free radical-mediated damage.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/physiopathology , Genes, Dominant , Genetic Predisposition to Disease , Islets of Langerhans/metabolism , Alloxan , Animals , Blood Glucose/metabolism , Body Weight , Crosses, Genetic , Diabetes Mellitus, Experimental/pathology , Female , Free Radicals/metabolism , Genotype , Glucose/pharmacology , Immunity, Innate , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred Strains , Sex Characteristics , Species Specificity , tert-Butylhydroperoxide/pharmacology
20.
Trends Biochem Sci ; 24(10): 390-3, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10500303

ABSTRACT

The identification of the fat mutation, which causes obesity in mice, as a defect in carboxypeptidase E (CPE) has raised more questions than answers. CPE is required for the processing of numerous neuroendocrine peptides and a mutation that inactivates CPE was predicted to be lethal. However, Cpe(fat) mutated mice live and become obese. So, why are mice with the Cpe(fat) mutation viable, and why does obesity develop as a consequence of the pleiotropic effects of this mutant allele? Recently, several new members of the carboxypeptidase family have been discovered, of which at least one, CPD, can partially compensate by contributing to neuroendocrine peptide processing. Obesity due to the Cpe(fat) mutation is not caused by increased food consumption but, rather, is a result of defective nutrient partitioning, the exact mechanism of which remains to be elucidated.


Subject(s)
Carboxypeptidases/genetics , Obesity/genetics , Peptides/metabolism , Animals , Carboxypeptidase H , Infertility/genetics , Obesity/enzymology , Obesity/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL