Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Genes Immun ; 17(7): 386-395, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27653816

ABSTRACT

Regulation of gene expression in immune cells is known to be under genetic control, and likely contributes to susceptibility to autoimmune diseases such as multiple sclerosis (MS). How this occurs in concert across multiple immune cell types is poorly understood. Using a mouse model that harnesses the genetic diversity of wild-derived mice, more accurately reflecting genetically diverse human populations, we provide an extensive characterization of the genetic regulation of gene expression in five different naive immune cell types relevant to MS. The immune cell transcriptome is shown to be under profound genetic control, exhibiting diverse patterns: global, cell-specific and sex-specific. Bioinformatic analysis of the genetically controlled transcript networks reveals reduced cell type specificity and inflammatory activity in wild-derived PWD/PhJ mice, compared with the conventional laboratory strain C57BL/6J. Additionally, candidate MS-GWAS (genome-wide association study candidate genes for MS susceptibility) genes were significantly enriched among transcripts overrepresented in C57BL/6J cells compared with PWD. These expression level differences correlate with robust differences in susceptibility to experimental autoimmune encephalomyelitis, the principal model of MS, and skewing of the encephalitogenic T-cell responses. Taken together, our results provide functional insights into the genetic regulation of the immune transcriptome, and shed light on how this in turn contributes to susceptibility to autoimmune disease.


Subject(s)
Autoimmunity/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Gene Expression Regulation , Genetic Variation , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Animals , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Genome-Wide Association Study , Male , Mice , Mice, Inbred C57BL , Sex Factors , Transcriptome
2.
Heredity (Edinb) ; 112(5): 508-18, 2014 May.
Article in English | MEDLINE | ID: mdl-24569637

ABSTRACT

External ear hole closure in LG/J mice represents a model of regenerative response. It is accompanied by the formation of a blastema-like structure and the re-growth of multiple tissues, including cartilage. The ability to regenerate tissue is heritable. An F34 advanced intercross line of mice (Wustl:LG,SM-G34) was generated to identify genomic loci involved in ear hole closure over a 30-day healing period. We mapped 19 quantitative trait loci (QTL) for ear hole closure. Individual gene effects are relatively small (0.08 mm), and most loci have co-dominant effects with phenotypically intermediate heterozygotes. QTL support regions were limited to a median size of 2 Mb containing a median of 19 genes. Positional candidate genes were evaluated using differential transcript expression between LG/J and SM/J healing tissue, function analysis and bioinformatic analysis of single-nucleotide polymorphisms in and around positional candidate genes of interest. Analysis of the set of 34 positional candidate genes and those displaying expression differences revealed over-representation of genes involved in cell cycle regulation/DNA damage, cell migration and adhesion, developmentally related genes and metabolism. This indicates that the healing phenotype in LG/J mice involves multiple physiological mechanisms.


Subject(s)
Chromosome Mapping/methods , Ear, External/physiology , Quantitative Trait Loci/genetics , Regeneration/genetics , Animals , Crosses, Genetic , Genotype , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Kinesins/genetics , Kinesins/metabolism , Mice , Mice, Inbred Strains , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Transcriptome/genetics , Wnt3A Protein/genetics , Wnt3A Protein/metabolism , Wound Healing/genetics
3.
Genes Immun ; 15(3): 168-75, 2014.
Article in English | MEDLINE | ID: mdl-24452267

ABSTRACT

Genetic studies of type 1 diabetes (T1D) have been advanced by comparative analysis of multiple susceptible and resistant rat strains with a permissive class II MHC haplotype, RT1(u). LEW.1WR1 (but not resistant LEW.1W or WF) rats are susceptible to T1D induced by a TLR3 agonist polyinosinic:polycytidylic acid followed by infection with parvovirus. We have mapped genetic loci for virus-induced T1D susceptibility, identifying a major susceptibility locus (Iddm37) near the MHC. The Iddm37 homologs on mouse and human chromosomes are also diabetes linked. We report that a major effect gene within Iddm37 is diubiquitin (Ubd). Gene expression profiling of pancreatic lymph nodes in susceptible and resistant rats during disease induction showed differences in Ubd transcript abundance. The LEW.1WR1 Ubd promoter allele leads to higher inducible levels of UBD than that of LEW.1W or WF. Using zinc-finger nucleases , we deleted a segment of the LEW.1WR1 Ubd gene and eliminated its expression. UBD-deficient rats show substantially reduced diabetes after viral infection. Complementary studies show that there may be another diabetes gene in addition to Ubd in the Iddm37 interval. These data prove that Ubd is a diabetes susceptibility gene, providing insight into the interplay of multiple genes and environmental factors in T1D susceptibility.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/virology , Genetic Predisposition to Disease , Parvovirinae , Ubiquitins/genetics , Alleles , Animals , Diabetes Mellitus, Type 1/mortality , Disease Models, Animal , Disease Susceptibility , Gene Expression , Gene Expression Profiling , Genetic Complementation Test , Genotype , Mice, Knockout , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Rats
4.
Genes Immun ; 14(6): 387-400, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23739610

ABSTRACT

The dilute plasma cytokine milieu associated with type 1 diabetes (T1D), while difficult to measure directly, is sufficient to drive transcription in a bioassay that uses healthy leukocytes as reporters. Previously, we reported disease-associated, partially IL-1 dependent, transcriptional signatures in both T1D patients and the BioBreeding (BB) rat model. Here, we examine temporal signatures in congenic BBDR.lyp/lyp rats that develop spontaneous T1D, and BBDR rats where T1D progresses only after immunological perturbation in young animals. After weaning, the BBDR temporal signature showed early coincident induction of transcription related to innate inflammation as well as IL-10- and TGF-ß-mediated regulation. BBDR plasma cytokine levels mirrored the signatures showing early inflammation, followed by induction of a regulated state that correlated with failure of virus to induce T1D in older rats. In contrast, the BBDR.lyp/lyp temporal signature exhibited asynchronous dynamics, with delayed induction of inflammatory transcription and later, weaker induction of regulatory transcription, consistent with their deficiency in regulatory T cells. Through longitudinal analyses of plasma-induced signatures in BB rats and a human T1D progressor, we have identified changes in immunoregulatory processes that attenuate a preexisting innate inflammatory state in BBDR rats, suggesting a mechanism underlying the decline in T1D susceptibility with age.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Disease Resistance , Transcriptome , Age Factors , Animals , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/virology , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Parvovirus , Rats , Rats, Inbred Strains , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
5.
Heredity (Edinb) ; 108(4): 441-6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22126848

ABSTRACT

Inbred mouse strains MRL and LG share the ability to fully heal ear hole punches with the full range of appropriate tissues without scarring. They also share a common ancestry, MRL being formed from a multi-strain cross with two final backcrosses to LG before being inbred by brother-sister mating. Many gene-mapping studies for healing ability have been performed using these two strains, resulting in the location of about 20 quantitative trait loci (QTLs). Here, we combine two of these crosses (N = 638), MRL/lpr × C57BL/6NTac and LG/J × SM/J, in a single combined cross analysis to increase the mapping power, decrease QTL support intervals, separate multiple QTLs and establish allelic states at individual QTL. The combined cross analysis located 11 QTLs, 6 affecting only one cross (5 LG × SM and 1 MRL × B6) and 5 affecting both crosses, approximately the number of common QTLs expected given strain SNP similarity. Amongst the five QTLs mapped in both crosses, three had significantly different genetic effects, additive in one cross and over or underdominant in the other. It is possible that allelic states at these three loci are different in SM and B6 because they lead to differences in dominance interactions with the LG and MRL alleles. QTL support intervals are 40% smaller in the combined cross analysis than in either of the single crosses. Combined cross analysis was successful in enhancing the interpretation of earlier QTL results for these strains.


Subject(s)
Alleles , Quantitative Trait Loci/genetics , Wound Healing/genetics , Animals , Crosses, Genetic , Female , Genotype , Lod Score , Male , Mice , Mice, Inbred Strains/genetics
6.
Genes Immun ; 11(8): 649-59, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20861860

ABSTRACT

Theiler's murine encephalomyelitis virus-induced demyelination (TMEVD) and experimental allergic encephalomyelitis (EAE) are the principal animal models of multiple sclerosis (MS). Previously, we provided evidence that Tmevd2 and Eae3 may represent either a shared susceptibility locus or members of a gene complex controlling susceptibility to central nervous system inflammatory demyelinating disease. To explore the genetic relationship between Tmevd2 and Eae3, we generated a D2.C-Tmevd2 interval-specific congenic (ISC) line and three overlapping interval-specific recombinant congenic (ISRC) lines in which the Tmevd2-resistant allele from BALB/cByJ was introgressed onto the TMEVD-susceptible DBA/2J background. These mice, all H2(d), were studied for susceptibility to EAE elicited by immunization with proteolipid protein peptide 180-199. Compared with DBA/2J mice, D2.C-Tmevd2 mice developed a significantly less severe clinical disease course and EAE pathology in the spinal cord, confirming the existence of Eae3 and its linkage to Tmevd2 in this strain combination. Compared with DBA/2J and D2.C-Tmevd2, all three ISRC lines exhibited clinical disease courses of intermediate severity. Neither differences in ex vivo antigen-specific cytokine nor proliferative responses uniquely cosegregated with differences in disease severity. These results indicate that multiple quantitative trait loci (QTLs) within the Tmevd2/Eae3 interval influence EAE severity, one of which includes a homology region for a QTL found in MS by admixture mapping.


Subject(s)
Animals, Genetically Modified/genetics , Demyelinating Diseases/genetics , Demyelinating Diseases/virology , Encephalomyelitis, Autoimmune, Experimental/genetics , Genetic Predisposition to Disease/genetics , Quantitative Trait Loci/genetics , Animals , Animals, Genetically Modified/immunology , Animals, Genetically Modified/virology , Cardiovirus Infections/genetics , Chromosomes, Mammalian/genetics , Demyelinating Diseases/immunology , Disease Models, Animal , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Genetic Markers , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Myelin Proteolipid Protein/immunology , Peptide Fragments/immunology , Theilovirus
7.
Physiol Genomics ; 38(1): 89-97, 2009 Jun 10.
Article in English | MEDLINE | ID: mdl-19351909

ABSTRACT

Congenic DRF.(f/f) rats are protected from type 1 diabetes (T1D) by 34 Mb of F344 DNA introgressed proximal to the gimap5 lymphopenia gene. To dissect the genetic factor(s) that confer protection from T1D in the DRF.(f/f) rat line, DRF.(f/f) rats were crossed to inbred BBDR or DR.(lyp/lyp) rats to generate congenic sublines that were genotyped and monitored for T1D, and positional candidate genes were sequenced. All (100%) DR.(lyp/lyp) rats developed T1D by 83 days of age. Reduction of the DRF.(f/f) F344 DNA fragment by 26 Mb (42.52-68.51 Mb) retained complete T1D protection. Further dissection revealed that a 2 Mb interval of F344 DNA (67.41-70.17 Mb) (region 1) resulted in 47% protection and significantly delayed onset (P < 0.001 compared with DR.(lyp/lyp)). Retaining <1 Mb of F344 DNA at the distal end (76.49-76.83 Mb) (region 2) resulted in 28% protection and also delayed onset (P < 0.001 compared with DR.(lyp/lyp)). Comparative analysis of diabetes frequency in the DRF.(f/f) congenic sublines further refined the RNO4 region 1 interval to approximately 670 kb and region 2 to the 340 kb proximal to gimap5. All congenic DRF.(f/f) sublines were prone to low-grade pancreatic mononuclear cell infiltration around ducts and vessels, but <20% of islets in nondiabetic rats showed islet infiltration. Coding sequence analysis revealed TCR Vbeta 8E, 12, and 13 as candidate genes in region 1 and znf467 and atp6v0e2 as candidate genes in region 2. Our results show that spontaneous T1D is controlled by at least two genetic loci 7 Mb apart on rat chromosome 4.


Subject(s)
Diabetes Mellitus, Experimental/genetics , GTP-Binding Proteins/genetics , Lymphopenia/genetics , Animals , Rats
8.
J Neurol Sci ; 236(1-2): 1-7, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16024047

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a progressive neuromuscular disorder. While most cases of ALS are sporadic, 10-15% are familial, and of these 15-20% possess a mutation in the gene that codes for the enzyme Cu/Zn superoxide dismutase (SOD1). In families of ALS patients with specific SOD1 mutations, affected members demonstrate significant heterogeneity of disease and a large variation in age of onset and severity, suggesting that there are genetic modifiers of disease expression. Transgenic mice expressing mutant forms of SOD1 demonstrate symptoms similar to those seen in patients with ALS. We have observed in our colony of G93A SOD1 transgenic mice a milder phenotype in mice in a C57BL/6J background than the C57BL/6JxSJL/J hybrid background used by Jackson Laboratories to maintain their colony. To investigate the effect of genetic background on phenotype, we have constructed congenic lines on two genetic backgrounds, C57BL/6J (B6) and SJL/J (SJL). We report the influence of background and gender on the survival of these congenic lines compared to the hybrid C57BL/6JxSJL/J background. The mean survival of G93A SOD1 mice in the hybrid B6/SJL background was 130 days, with females surviving significantly longer than males. When compared to the hybrid B6/SJL background, the survival of mice in the SJL background significantly decreased, and the gender difference in survival was maintained. On the other hand, mean survival in the B6 background significantly increased, and in contrast to the B6/SJL and SJL backgrounds, there was no difference in survival between males and females. Transgene copy numbers were verified in all animals to ensure that any phenotypic differences observed were not due to alterations in copy number. This is the first report of a shortened lifespan when the G93A SOD1 transgene is placed on the SJL/J background and an increased survival with the loss of gender influences when the transgene is placed on the C57BL/6J background.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/mortality , Disease Models, Animal , Sex Characteristics , Age Factors , Animals , Genotype , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Superoxide Dismutase/genetics , Survival
9.
Am J Pathol ; 157(2): 637-45, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10934166

ABSTRACT

Experimental allergic encephalomyelitis (EAE) is the principal genetically determined animal model for multiple sclerosis (MS), the major inflammatory disease of the central nervous system (CNS). Although genetics clearly play a role in susceptibility to MS, attempts to identify the underlying genes have been disappointing. Considerable variation exists between MS patients with regard to the severity of clinical signs, mechanism of demyelination, and location of CNS lesions, confounding the interpretation of genetic data. A mouse-human synteny mapping approach may allow the identification of candidate susceptibility loci for MS based on the location of EAE susceptibility loci. To date, 16 regions of the mouse genome have been identified that control susceptibility or clinical signs of EAE. In this work, we examined the genetic control of histopathological lesions of EAE in an F2 intercross population generated from the EAE susceptible SJL/J and EAE resistant B10.S/DvTe mouse strains. Composite interval mapping was used to identify 10 quantitative trait loci (QTL), including seven newly identified loci controlling the distribution and severity of CNS lesions associated with murine EAE. QTL on chromosome 10 control lesions in the brain, whereas QTL on chromosomes 3, 7, and 12 control lesions in the spinal cord. Furthermore, sexually dimorphic QTL on chromosomes 2, 9, and 11 control CNS lesions in females, whereas QTL on chromosomes 10, 11, 12, 16, and 19 control lesions in males. Our results suggest that the severity and location of CNS lesions in EAE are genetically controlled, and that the genetic component controlling the character and severity of the lesions can be influenced by sex.


Subject(s)
Brain/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Quantitative Trait, Heritable , Spinal Cord/metabolism , Animals , Brain/pathology , Central Nervous System/metabolism , Central Nervous System/pathology , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Inflammation/genetics , Inflammation/pathology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Male , Mice , Mice, Inbred Strains , Microsatellite Repeats , Severity of Illness Index , Sex Factors , Spinal Cord/pathology
10.
J Immunol ; 164(6): 3420-5, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10706738

ABSTRACT

Pertussis toxin (PTX) is a potent ancillary adjuvant used to elicit several different autoimmune diseases, including experimental allergic encephalomyelitis (EAE). To delineate the genetics of PTX effect in EAE, we mapped EAE-modifying (eae-m) loci in cohorts of backcross mice immunized with and without PTX. In this study, we analyzed the genetic basis of EAE susceptibility and severity and the intermediate phenotypes of mononuclear cell infiltration, suppuration, and demyelination. In animals immunized with PTX, one major locus, eae9, controls disease susceptibility and severity. Eae9 also regulates the extent of mononuclear cell infiltration of the spinal cord in male mice. Without PTX, five eae-m loci were noted, including three new loci in intervals on chromosomes 8 (eae14), 10 (eae17), and 18 (eae18). Taken together, these results suggest that eae9 controls the effects of PTX in EAE susceptibility, and is capable of overriding the other genetic checkpoints in the pathogenesis of this disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Genetic Predisposition to Disease/genetics , Pertussis Toxin , Virulence Factors, Bordetella/immunology , Animals , Brain/pathology , Crosses, Genetic , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Genetic Markers , Genetic Predisposition to Disease/etiology , Histamine/immunology , Linear Models , Male , Mice , Mice, Inbred C57BL , Quantitative Trait, Heritable , Severity of Illness Index , Spinal Cord/pathology , Virulence Factors, Bordetella/toxicity
11.
J Cell Biochem ; 77(1): 1-5, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10679811

ABSTRACT

Puralpha is a 39-kDa sequence-specific single-stranded DNA/RNA binding protein with the ability to modulate transcription of several genes containing the Pur element in their promoter region. Human and mouse Puralpha exhibit an extraordinary degree of conservation with only two changes at amino acid residues 49 and 306. A 15-kb genomic clone encompassing the mouse Puralpha gene was isolated by screening the mouse genomic library, using a PCR-amplified fragment from human Puralpha cDNA. Results from sequencing analysis confirmed the isolated genomic clone to be Puralpha and not the other members of the Pur family, including Purbeta. Characterization of the mouse Puralpha gene by restriction analysis/Southern blotting and sequencing revealed that the Puralpha gene contains only one intron within the 5' UTR and the open reading frame was intact. Using chromosomal markers, the Puralpha gene was mapped to chromosome 18 in mouse and rat.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Animals , Chromosome Mapping , Cloning, Molecular , DNA-Binding Proteins , Humans , Mice , Nerve Tissue Proteins , Rats , Sequence Analysis , Transcription Factors
12.
J Virol ; 73(11): 9232-6, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10516031

ABSTRACT

Certain inbred mouse strains display progression to lymphoma development after infection with E-55+ murine leukemia virus (E-55+ MuLV), while others demonstrate long-term nonprogression. This difference in disease progression occurs despite the fact that E-55+ MuLV causes persistent infection in both immunocompetent BALB/c-H-2(k) (BALB.K) progressor (P) and C57BL/10-H-2(k) (B10.BR) long-term nonprogressor (LTNP) mice. In contrast to immunocompetent mice, immunosuppressed mice from both P and LTNP strains develop lymphomas about 2 months after infection, indicating that the LTNP phenotype is determined by the immune response of the infected mouse. In this study, we used bone marrow chimeras to demonstrate that the LTNP phenotype is associated with the genotype of donor bone marrow and not the recipient microenvironment. In addition, we have mapped a genetic locus that may be responsible for the LTNP trait. Microsatellite-based linkage analysis demonstrated that a non-major histocompatibility complex gene on chromosome 15 regulates long-term survival and is located in the same region as the Rfv3 gene. Rfv3 is involved in recovery from Friend virus-induced leukemia and has been demonstrated to regulate neutralizing virus antibody titers. In our studies, however, both P and LTNP strains produce similar titers of neutralizing and cytotoxic anti-E-55+ MuLV. Therefore, while it is possible that Rfv3 influences the course of E-55+ MuLV infection, it is more likely that the LTNP phenotype in E-55+ MuLV-infected mice is regulated by a different, closely linked gene.


Subject(s)
Leukemia Virus, Murine/immunology , Leukemia, Experimental/genetics , Retroviridae Infections/genetics , Animals , Bone Marrow Cells , Disease Progression , Disease Susceptibility , Genetic Linkage , Immunity, Innate , Inbreeding , Leukemia, Experimental/immunology , Leukemia, Experimental/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phenotype , Retroviridae Infections/immunology , Retroviridae Infections/virology , Tumor Virus Infections/genetics , Tumor Virus Infections/immunology , Tumor Virus Infections/virology
13.
Diabetes ; 48(11): 2138-44, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10535446

ABSTRACT

Diabetes-prone (DP) BB rats develop autoimmune type 1 diabetes spontaneously. At least five loci are linked to disease expression: the major histocompatibility complex (iddm2), two susceptibility loci (iddm4, iddm5), and, possibly, a resistance locus (iddm3). Spontaneous disease also requires homozygosity for lyp/iddm1, which causes lymphopenia. It has not been determined whether lyp/iddm1 is required for predisposition to diabetes autoimmunity in addition to being required for its spontaneous expression. We analyzed backcross rats segregating for diabetes but not lymphopenia using Wistar-Furth (WF) and diabetes-resistant (DR) BB animals. The latter are nonlymphopenic (lyp+/+) and develop diabetes only in response to immunological perturbants. Treatment of (DR-BB x WF)F1 x WF animals (all lyp+/+) using a standard induction protocol caused type 1 diabetes in 58% of progeny. Expression of type 1 diabetes was strongly linked to iddm4. The results suggest that lyp/iddm1 does not determine the predisposition to autoimmunity in BB rats and that iddm4 is a major diabetogenic locus in both DP- and DR-BB animals. The iddm4 gene maps to a region containing several major autoimmunity loci, including aia2, aia3, and cia3. We propose that BB rat diabetes requires 1) class II RT1u (iddm2) for susceptibility, 2) additional loci for disease initiation and progression in response to perturbants, and 3) lyp for spontaneous disease.


Subject(s)
Chromosome Mapping , Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease/genetics , Rats, Inbred BB/genetics , Age of Onset , Animals , Crosses, Genetic , Diabetes Mellitus, Type 1/physiopathology , Genetic Markers , Homozygote , Immunity, Innate/genetics , Lod Score , Lymphopenia/genetics , Microsatellite Repeats , Polymerase Chain Reaction , Rats , Species Specificity
14.
J Immunol ; 163(4): 2262-6, 1999 Aug 15.
Article in English | MEDLINE | ID: mdl-10438970

ABSTRACT

Experimental allergic encephalomyelitis (EAE), the principal animal model of multiple sclerosis, is genetically controlled. To date, 13 disease-modifying loci have been identified in the mouse by whole genome scanning using an F2 intercross between EAE-susceptible SJL/J and EAE-resistant B10.S/DvTe mice. Two quantitative trait loci (QTL), eae6 and eae7, on chromosome 11 were identified by classical marker-specific linkage analysis and interval mapping. Both QTL were reported to be associated with severity and duration of clinical signs. eae7 was subsequently shown to be a unique locus controlling the development of monophasic remitting/nonrelapsing EAE. In this study, composite interval mapping resolved eae6 into two linked QTL: eae6a at 0-13 cM is associated with disease severity, and eae6b at 19-28 cM associated with the duration of clinical signs. Additionally, composite interval mapping significantly refined the locations of eae6a, eae6b, and eae7, thereby facilitating systematic candidate gene screening by cDNA sequencing of SJL/J and B10.S/DvTe alleles. Sequence polymorphisms were not seen in Lif and IL12 beta, candidate genes for eae6a and eae6b, respectively. Similarly, cDNA sequence polymorphisms in Nos2, Scya3, Scya4, Scya5, Scya6, Scya7, Scya9, Scya10, and Scya11 were excluded as candidates for eae7. However, multiple sequence polymorphisms resulting in significant amino acid substitutions were identified in Scya1 (TCA-3), Scya2 (monocyte chemoattractant protein (MCP)-1), and Scya12 (MCP-5). Given the role of chemokines in EAE, these sequence polymorphisms are promising candidates for eae7, a locus associated with severity of clinical signs and susceptibility to the shorter, less severe monophasic remitting/nonrelapsing form of disease.


Subject(s)
Chemokines, CC , Chemokines/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Genetic Markers , Polymorphism, Genetic/immunology , Amino Acid Sequence , Animals , Chemokine CCL1 , Chemokine CCL2/genetics , Chromosomes, Human, Pair 11/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Genes, Overlapping/immunology , Genetic Predisposition to Disease/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Molecular Sequence Data , Monocyte Chemoattractant Proteins/genetics , Quantitative Trait, Heritable
15.
J Immunol ; 162(5): 3096-102, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10072563

ABSTRACT

Experimental allergic encephalomyelitis (EAE) is the principal animal model of multiple sclerosis (MS), the major inflammatory disease of the central nervous system. Murine EAE is generally either an acute monophasic or relapsing disease. Because the clinical spectrum of MS is more diverse, the limited range of disease subtypes observed in EAE has raised concern regarding its relevance as a model for MS. During the generation of a large F2 mapping population between the EAE-susceptible SJL/J and EAE-resistant B10.S/DvTe inbred lines, we identified four distinct subtypes of murine EAE resembling clinical subtypes seen in MS. We observed acute progressive, chronic/nonremitting, remitting/relapsing, and monophasic remitting/nonrelapsing EAE. An additional subtype, benign EAE, was identified after histologic examination revealed that some mice had inflammatory infiltrates of the central nervous system, but did not show clinical signs of EAE. Genome exclusion mapping was performed to identify the loci controlling susceptibility to each disease subtype. We report three novel EAE-modifying loci on chromosomes 16, 7, and 13 (eae11-13, respectively). Additionally, unique loci with gender-specific effects govern susceptibility to remitting/relapsing (eae12) and monophasic remitting/nonrelapsing (eae7 and 13) EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Sex Characteristics , Animals , Chromosome Mapping , Encephalomyelitis, Autoimmune, Experimental/classification , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Genetic Linkage , Genetic Predisposition to Disease , Male , Mice , Recurrence
16.
Diabetes ; 48(1): 50-8, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9892222

ABSTRACT

BB rats are used as models of autoimmune human IDDM. Genetic control of IDDM in both species is complex, including both major histocompatibility complex (MHC)-linked and non-MHC-linked genes. DP-BB rats develop IDDM spontaneously. Expression of disease in these animals requires homozygosity at the lyp locus, which causes lymphopenia. All genetic analyses of BB rat diabetes to date have backcrossed to the DP-BB strain or used (DP-BB x non-BB)F2 animals to ensure that a fraction of progeny are homozygous for lyp. Here we report the analysis of a backcross of the DP-BB rat to the histocompatible WF rat. Neither WF nor (WF x DP-BB)F1 animals develop spontaneous IDDM. However, 95% of (WF x DP-BB)F1 rats and a fraction of (WF x DP-BB) x WF backcross animals readily develop IDDM after treatment with polyinosinic:polycytidylic acid and a cytotoxic anti-RT6.1 monoclonal antibody. Using simple sequence length polymorphism analysis, we have mapped loci on chromosomes 4 and 13 that show significant linkage to IDDM expression and insulitis. The susceptibility locus on chromosome 4 is linked to, but not identical to, lyp. We propose a disease model for the BB rat that requires 1) the RT1u MHC haplotype for disease susceptibility, 2) a new locus on chromosome 4 for disease initiation (as measured by insulitis), 3) a new locus on chromosome 13 for disease progression in response to environmental perturbation, and 4) lyp for spontaneous expression of disease.


Subject(s)
Chromosomes, Human, Pair 13/genetics , Chromosomes, Human, Pair 4/genetics , Diabetes Mellitus, Type 1/genetics , Genetic Linkage/drug effects , Genetic Predisposition to Disease/genetics , Major Histocompatibility Complex/genetics , Animals , Chromosome Mapping , Diabetes Mellitus, Type 1/etiology , Disease Progression , Genome , Humans , Hybridization, Genetic , Islets of Langerhans , Pancreatitis/complications , Pancreatitis/genetics , Pancreatitis/physiopathology , Phenotype , Rats , Rats, Inbred BB/genetics , Rats, Inbred WF/genetics
17.
Proc Natl Acad Sci U S A ; 95(20): 11792-7, 1998 Sep 29.
Article in English | MEDLINE | ID: mdl-9751744

ABSTRACT

Wound healing of mammalian tissue is an essential process in the maintenance of body integrity. The general mechanism of wound healing usually studied in adult mammals is repair, in contrast to the regeneration seen in more primitive vertebrates. We recently have discovered that MRL/MpJ mice, unlike all other strains of mice tested, undergo rapid and complete wound closure that resembles regeneration. Specifically, through-and-through surgical ear hole wounds close without scarring in <4 weeks with normal gross and microanatomic architecture, including chondrogenesis. We also demonstrated that this healing is a heritable trait in inbred mice. In this study, we present results pertaining to its genetic control in progeny segregating for this phenotype. To identify the genetic loci that control the wound closure process, a genome-wide scan was performed on (MRL/MpJ-Faslpr x C57BL/6)F2 and backcross populations. In the primary screens of these populations, quantitative trait loci that control the extent of wound closure were detected on chromosomes 8, 12, and 15 and at two separate locations on chromosome 13. Evidence of further genetic control of healing was found on chromosome 7. All alleles that contribute to full wound closure are derived from the MRL/MpJ-Faslpr parent except for the quantitative trait locus on chromosome 8, which is derived from C57BL/6.


Subject(s)
Wound Healing/genetics , Alleles , Animals , Chromosome Mapping , Crosses, Genetic , Ear/injuries , Ear/physiology , Female , Genetic Markers , Male , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Phenotype , Polymorphism, Genetic , Quantitative Trait, Heritable , Regeneration/genetics
18.
J Immunol ; 161(4): 1860-7, 1998 Aug 15.
Article in English | MEDLINE | ID: mdl-9712054

ABSTRACT

Experimental allergic encephalomyelitis (EAE), the principal animal model of multiple sclerosis, is a genetically determined phenotype. In this study, analyses of the cumulative disease frequencies in parental, F1 hybrid, and F2 mice, derived from the EAE-susceptible SJL/J strain and the EAE-resistant B10.S/DvTe strain, confirmed that susceptibility to EAE is not inherited as a simple Mendelian trait. Whole genome scanning, using 150 informative microsatellite markers and a panel of 291 affected and 390 unaffected F2 progeny, revealed significant linkage of EAE susceptibility to marker loci on chromosomes 7 (eae4) and 17, distal to H2 (eae5). Quantitative trait loci for EAE severity, duration, and onset were identified on chromosomes 11 (eae6, and eae7), 2 (eae8), 9 (eae9), and 3 (eae10). While each locus reported in this study is important in susceptibility or disease course, interactions between marker loci were not statistically significant in models of genetic control. One locus, eae7, colocalizes to the same region of chromosome II as Orch3 and Idd4, susceptibility loci in autoimmune orchitis and insulin-dependent diabetes mellitus, respectively. Importantly, eae5 and eae7 are syntenic with human chromosomes 6p21 and 17q22, respectively, two regions of potential significance recently identified in human multiple sclerosis genome scans.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Quantitative Trait, Heritable , Animals , Chromosome Mapping , Crosses, Genetic , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/epidemiology , Female , Genetic Markers/immunology , Humans , Incidence , Linear Models , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology
19.
J Immunol ; 161(1): 17-26, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9647202

ABSTRACT

E-55+ murine leukemia virus infection of both progressor (BALB) and long term nonprogressor (C57BL) mouse strains is characterized by an acute and a persistent phase of infection. During the acute phase, progressor strains require CD8+ T cells to decrease virus burden, whereas the long term nonprogressor strains do not. In the present studies the immune response in BALB and C57BL mice during the acute phase of E-55+ murine leukemia virus infection was examined. The results demonstrate that BALB mice produce both IL-4 and IFN-gamma, in contrast to C57BL mice, which produce only IFN-gamma. In BALB mice, IL-4 production results in the absolute requirement for CD8+ T cells to reduce the virus burden during the acute phase of infection. The anti-virus immune response in these mice is IFN-gamma dependent. On the other hand, C57BL mice do not produce IL-4 and, in the absence of both CD8+ T cells and IFN-gamma, still generate an effective anti-virus immune response. Genetic studies suggest that these distinct immune responses are regulated by more than one non-MHC-linked gene. Two candidate regions that may encode this gene(s), located on chromosomes 7 and 19, respectively, were identified by recombinant inbred strain linkage analysis.


Subject(s)
Leukemia Virus, Murine/immunology , Retroviridae Infections/immunology , Tumor Virus Infections/immunology , Acute Disease , Animals , Cytokines/biosynthesis , Disease Susceptibility , Genetic Linkage , Immunity, Innate , Interferon-gamma/physiology , Interleukin-4/biosynthesis , Interleukin-4/physiology , Leukemia, Experimental/genetics , Leukemia, Experimental/immunology , Leukemia, Experimental/metabolism , Major Histocompatibility Complex/genetics , Major Histocompatibility Complex/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Retroviridae Infections/genetics , Retroviridae Infections/metabolism , Tumor Virus Infections/genetics , Tumor Virus Infections/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...