Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Genome Res ; 23(9): 1474-85, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23800453

RESUMEN

Understanding the DNA elements that constitute and control the regulatory genome is critical for the appropriate therapeutic management of complex diseases. Here, using chromosome Y (ChrY) consomic mouse strains on the C57BL/6J (B6) background, we show that susceptibility to two diverse animal models of autoimmune disease, experimental allergic encephalomyelitis (EAE) and experimental myocarditis, correlates with the natural variation in copy number of Sly and Rbmy multicopy ChrY genes. On the B6 background, ChrY possesses gene regulatory properties that impact genome-wide gene expression in pathogenic CD4(+) T cells. Using a ChrY consomic strain on the SJL background, we discovered a preference for ChrY-mediated gene regulation in macrophages, the immune cell subset underlying the EAE sexual dimorphism in SJL mice, rather than CD4(+) T cells. Importantly, in both genetic backgrounds, an inverse correlation exists between the number of Sly and Rbmy ChrY gene copies and the number of significantly up-regulated genes in immune cells, thereby supporting a link between copy number variation of Sly and Rbmy with the ChrY genetic element exerting regulatory properties. Additionally, we show that ChrY polymorphism can determine the sexual dimorphism in EAE and myocarditis. In humans, an analysis of the CD4(+) T cell transcriptome from male multiple sclerosis patients versus healthy controls provides further evidence for an evolutionarily conserved mechanism of gene regulation by ChrY. Thus, as in Drosophila, these data establish the mammalian ChrY as a member of the regulatory genome due to its ability to epigenetically regulate genome-wide gene expression in immune cells.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Encefalomielitis Autoinmune Experimental/genética , Predisposición Genética a la Enfermedad , Macrófagos/metabolismo , Miocarditis/genética , Transcriptoma , Cromosoma Y/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular , Animales , Variaciones en el Número de Copia de ADN , Femenino , Dosificación de Gen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Caracteres Sexuales
2.
Proc Natl Acad Sci U S A ; 107(44): 18967-72, 2010 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-20956310

RESUMEN

Disruption of the blood-brain barrier (BBB) underlies the development of experimental autoimmune encephalomyelitis (EAE) and multiple sclerosis. Environmental factors, such as Bordetella pertussis, are thought to sensitize central endothelium to biogenic amines like histamine, thereby leading to increased BBB permeability. B. pertussis-induced histamine sensitization (Bphs) is a monogenic intermediate phenotype of EAE controlled by histamine H(1) receptor (Hrh1/H(1)R). Here, we transgenically overexpressed H(1)R in endothelial cells of Hrh1-KO (H(1)RKO) mice to test the role of endothelial H(1)R directly in Bphs and EAE. Unexpectedly, transgenic H(1)RKO mice expressing endothelial H(1)R under control of the von Willebrand factor promoter (H(1)RKO-vWF(H1R) Tg) were Bphs-resistant. Moreover, H(1)RKO-vWF(H1R) Tg mice exhibited decreased BBB permeability and enhanced protection from EAE compared with H(1)RKO mice. Thus, contrary to prevailing assumptions, our results show that endothelial H(1)R expression reduces BBB permeability, suggesting that endothelial H(1)R signaling may be important in the maintenance of cerebrovascular integrity.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Permeabilidad Capilar , Encefalomielitis Autoinmune Experimental/metabolismo , Endotelio Vascular/metabolismo , Receptores Histamínicos H1/metabolismo , Transducción de Señal , Animales , Bordetella pertussis/genética , Bordetella pertussis/metabolismo , Encefalomielitis Autoinmune Experimental/genética , Predisposición Genética a la Enfermedad , Ratones , Ratones Noqueados , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Receptores Histamínicos H1/genética , Tos Ferina/genética , Tos Ferina/metabolismo , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo
3.
Commun Biol ; 6(1): 244, 2023 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-36879097

RESUMEN

Histamine plays pivotal role in normal physiology and dysregulated production of histamine or signaling through histamine receptors (HRH) can promote pathology. Previously, we showed that Bordetella pertussis or pertussis toxin can induce histamine sensitization in laboratory inbred mice and is genetically controlled by Hrh1/HRH1. HRH1 allotypes differ at three amino acid residues with P263-V313-L331 and L263-M313-S331, imparting sensitization and resistance respectively. Unexpectedly, we found several wild-derived inbred strains that carry the resistant HRH1 allotype (L263-M313-S331) but exhibit histamine sensitization. This suggests the existence of a locus modifying pertussis-dependent histamine sensitization. Congenic mapping identified the location of this modifier locus on mouse chromosome 6 within a functional linkage disequilibrium domain encoding multiple loci controlling sensitization to histamine. We utilized interval-specific single-nucleotide polymorphism (SNP) based association testing across laboratory and wild-derived inbred mouse strains and functional prioritization analyses to identify candidate genes for this modifier locus. Atg7, Plxnd1, Tmcc1, Mkrn2, Il17re, Pparg, Lhfpl4, Vgll4, Rho and Syn2 are candidate genes within this modifier locus, which we named Bphse, enhancer of Bordetella pertussis induced histamine sensitization. Taken together, these results identify, using the evolutionarily significant diversity of wild-derived inbred mice, additional genetic mechanisms controlling histamine sensitization.


Asunto(s)
Bordetella pertussis , Histamina , Animales , Ratones , Bordetella pertussis/genética , Toxina del Pertussis , Transducción de Señal , Proteínas del Sistema Complemento , Sitios Genéticos , Glicoproteínas de Membrana , Péptidos y Proteínas de Señalización Intracelular , Ribonucleoproteínas
4.
Ann Neurol ; 70(6): 887-96, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22190363

RESUMEN

OBJECTIVE: The major histocompatibility complex (MHC) is the primary genetic contributor to multiple sclerosis (MS) and experimental allergic encephalomyelitis (EAE), but multiple additional interacting loci are required for genetic susceptibility. The identity of most of these non-MHC genes is unknown. In this report, we identify genes within evolutionarily conserved genetic pathways leading to MS and EAE. METHODS: To identify non-MHC binary and quantitative trait loci (BTL/QTL) important in the pathogenesis of EAE, we generated phenotype-selected congenic mice using EAE-resistant B10.S and EAE-susceptible SJL mice. We hypothesized that genes linked to EAE BTL/QTL and MS-GWAS can be identified if they belong to common evolutionarily conserved pathways, which can be identified with a bioinformatic approach using Ingenuity software. RESULTS: Many known BTL/QTL were retained and linked to susceptibility during phenotype selection, the most significant being a region on chromosome 17 distal to H2 (Eae5). We show in pathway analysis that T helper (T(H))-cell differentiation genes are critical for both diseases. Bioinformatic analyses predicted that Eae5 is important in CD4 T-effector and/or Foxp3(+) T-regulatory cells (Tregs), and we found that B10.S-Eae5(SJL) congenic mice have significantly greater numbers of lymph node CD4 and Tregs than B10.S mice. INTERPRETATION: These results support the polygenic model of MS/EAE, whereby MHC and multiple minor loci are required for full susceptibility, and confirm a critical genetic dependence on CD4 T(H)-cell differentiation and function in the pathogenesis of both diseases.


Asunto(s)
Susceptibilidad a Enfermedades , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Complejo Mayor de Histocompatibilidad/genética , Esclerosis Múltiple/genética , Linfocitos T Colaboradores-Inductores/patología , Animales , Linfocitos T CD4-Positivos/fisiología , Biología Computacional , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/etiología , Citometría de Flujo , Adyuvante de Freund/efectos adversos , Ligamiento Genético , Estudio de Asociación del Genoma Completo , Ratones , Ratones Congénicos , Fenotipo , Sitios de Carácter Cuantitativo/genética , Estadísticas no Paramétricas
5.
J Immunol ; 182(12): 7776-83, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19494301

RESUMEN

Genes controlling immunopathologic diseases of differing etiopathology may also influence susceptibility to autoimmune disease. B10.D1-H2(q)/SgJ mice with a 2538 G-->A missense mutation in the tyrosine kinase-2 gene (Tyk2) are susceptible to Toxoplasma gondii yet resistant to autoimmune arthritis, unlike the wild-type B10.Q/Ai substrain. To understand whether Tyk2 is also important in a second autoimmune model, experimental allergic encephalomyelitis (EAE) was induced in B10.D1-H2(q)/SgJ (Tyk2(A)) and B10.Q/Ai (Tyk2(G)) mice with the myelin oligodendrocyte glycoprotein peptide 79-96. B10.D1-H2(q)/SgJ mice were resistant to EAE whereas B10.Q/Ai mice were susceptible, and a single copy of the Tyk2(G) allele conferred EAE susceptibility in F(1) hybrids. Furthermore, EAE resistance in B10.D1-H2(q)/SgJ mice was overridden when pertussis toxin (PTX) was used to mimic the effects of environmental factors derived from infectious agents. Numerous cytokines and chemokines were increased when PTX was included in the immunization protocol. However, only RANTES, IL-6, and IFN-gamma increased significantly with both genetic compensation and PTX treatment. These data indicate that Tyk2 is a shared autoimmune disease susceptibility gene whose genetic contribution to disease susceptibility can be modified by environmental factors. Single nucleotide polymorphisms like the one that distinguishes Tyk2 alleles are of considerable significance given the potential role of gene-by-environment interactions in autoimmune disease susceptibility.


Asunto(s)
Encefalomielitis Autoinmune Experimental/enzimología , Encefalomielitis Autoinmune Experimental/genética , Predisposición Genética a la Enfermedad/genética , Polimorfismo de Nucleótido Simple/genética , TYK2 Quinasa/genética , TYK2 Quinasa/metabolismo , Alelos , Animales , Células Cultivadas , Citocinas/biosíntesis , Citocinas/inmunología , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/patología , Ratones , Proteínas de la Mielina , Glicoproteína Asociada a Mielina/farmacología , Glicoproteína Mielina-Oligodendrócito , Toxina del Pertussis/farmacología
6.
J Immunol ; 182(4): 1789-93, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19201829

RESUMEN

Multiple sclerosis is a sexually dimorphic, demyelinating disease of the CNS, and experimental allergic encephalomyelitis (EAE) is its principal autoimmune model. Young male SJL/J mice are relatively resistant to EAE whereas older males and SJL/J females of any age are susceptible. By comparing a wide age range of proteolipid protein peptide 139-151 immunized mice, we found that female disease severity remains constant with age. In contrast, EAE disease severity increases with age in SJL/J males, with young males having significantly less severe disease and older males having significantly more disease than equivalently aged females. To determine whether the Y chromosome contributes to this sexual dimorphism, EAE was induced in consomic SJL/J mice carrying a B10.S Y chromosome (SJL.Y(B10.S)). EAE was significantly more severe in young male SJL.Y(B10.S) mice compared with young male SJL/J mice. These studies show that a Y chromosome-linked polymorphism controls the age-dependent EAE sexual dimorphism observed in SJL/J mice.


Asunto(s)
Encefalomielitis Autoinmune Experimental/genética , Predisposición Genética a la Enfermedad/genética , Caracteres Sexuales , Cromosoma Y/genética , Factores de Edad , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Masculino , Ratones , Estaciones del Año
7.
Genes (Basel) ; 12(6)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34205929

RESUMEN

Genetic analyses of human type 1 diabetes (T1D) have yet to reveal a complete pathophysiologic mechanism. Inbred rats with a high-risk class II major histocompatibility complex (MHC) haplotype (RT1B/Du) can illuminate such mechanisms. Using T1D-susceptible LEW.1WR1 rats that express RT1B/Du and a susceptible allele of the Ubd promoter, we demonstrate that germline knockout of Tcrb-V13S1A1, which encodes the Vß13a T cell receptor ß chain, completely prevents diabetes. Using the RT1B/Du-identical LEW.1W rat, which does not develop T1D despite also having the same Tcrb-V13S1A1 ß chain gene but a different allele at the Ubd locus, we show that knockout of the Ubash3a regulatory gene renders these resistant rats relatively susceptible to diabetes. In silico structural modeling of the susceptible allele of the Vß13a TCR and its class II RT1u ligand suggests a mechanism by which a germline TCR ß chain gene could promote susceptibility to T1D in the absence of downstream immunoregulation like that provided by UBASH3A. Together these data demonstrate the critical contribution of the Vß13a TCR to the autoimmune synapse in T1D and the regulation of the response by UBASH3A. These experiments dissect the mechanisms by which MHC class II heterodimers, TCR and regulatory element interact to induce autoimmunity.


Asunto(s)
Autoinmunidad/genética , Diabetes Mellitus Tipo 1/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Animales , Diabetes Mellitus Tipo 1/inmunología , Genotipo , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Insulina/química , Insulina/inmunología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Unión Proteica , Ratas , Ratas Endogámicas Lew , Receptores de Antígenos de Linfocitos T alfa-beta/química , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología
8.
J Control Release ; 331: 164-175, 2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33450320

RESUMEN

The development of nanomaterials to induce antigen-specific immune tolerance has shown promise for treating autoimmune diseases. While PEGylation has been widely used to reduce host immune responses to nanomaterials, its tolerogenic potential has not been reported. Here, we report for the first time that a subcutaneous injection of PEGylated poly(lactide-co-glycolide) (PLGA) nanoparticles containing auto-antigen peptide MOG35-55 without any tolerogenic drugs is sufficient to dramatically ameliorate symptoms after disease onset in an antigen-specific manner in a mouse model of multiple sclerosis. Neither free MOG35-55 nor particles without PEG exhibit this efficacy. Interestingly, mechanistic studies indicate that PEGylation of nanoparticles does not reduce dendritic cell activation through direct nanoparticle-cell interactions. Instead, PEGylated nanoparticles induce lower complement activation, neutrophil recruitment, and co-stimulatory molecule expression on dendritic cells around the injection sitecompared to non-PEGylated PLGA nanoparticles, creating a more tolerogenic microenvironment in vivo. We further demonstrate that the locally recruited dendritic cells traffic to lymphoid organs to induce T cell tolerance. These results highlight the critical role of surface properties of nanomaterials in inducing immune tolerance via subcutaneous administration.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Nanopartículas , Animales , Antígenos , Células Dendríticas , Tolerancia Inmunológica , Ratones
9.
Front Endocrinol (Lausanne) ; 12: 629242, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815287

RESUMEN

The depleting Vß13a T cell receptor monoclonal antibody (mAb) 17D5 prevents both induced and spontaneous autoimmune diabetes in BB rats. Here it was tested in congenic DRLyp/Lyp rats, all of which spontaneously developed diabetes. Starting at 40 days of age, rats were injected once weekly with either saline, His42 Vß16 mAb, or 17D5 mAb and monitored for hyperglycemia. Diabetes occurred in 100% (n = 5/5) of saline-treated rats (median age, 66 days; range 55-73), and in 100% (n = 6/6) of His42-treated rats (median age, 69 days; range 59-69). Diabetes occurred in fewer (n = 8/11, 73%) 17D5-treated rats at a later age (median 76 days, range 60-92). Three (27%) of the 17D5-treated rats were killed at 101-103 days of age without diabetes (17D5 no-diabetes rats). Survival analysis demonstrated that 17D5 mAb delayed diabetes onset. Saline- and His42-treated rats had severely distorted islets with substantial loss of insulin-positive cells. These rats exhibited prominent hyaluronan (HA) staining, with the intra-islet HA+ accumulations measuring 5,000 ± 2,400 µm2 and occupying 36 ± 12% of islet area, and severe (grade 4) insulitis with abundant infiltration by CD68+, CD3+, and CD8+ cells. The 17D5 mAb-treated rats with delayed diabetes onset exhibited less severe insulitis (predominantly grade 3). In contrast, the 17D5 no-diabetes rats had mostly normal islets, with insulin+ cells representing 76 ± 3% of islet cells. In these rats, the islet HA deposits were significantly smaller than in the diabetic rats; the intra-islet HA+ areas were 1,200 ± 300 µm2 and accounted for 8 ± 1% of islet area. Also, islet-associated CD68+ and CD3+ cells occurred less frequently (on average in 60 and 3% of the islets, respectively) than in the diabetes rats (present in >95% of the islets). No CD8+ cells were detected in islets in all 17D5 no-diabetes rats. We conclude that mAb 17D5 delayed diabetes in DRLyp/Lyp rats and markedly reduced expression of HA and concomitant infiltration of CD68+, CD3+, and CD8+ cells. Our findings underscore the importance of refining immune suppression in prevention or intervention clinical trials to use mAb reagents that are directed against specific T cell receptors.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Complejo CD3/metabolismo , Linfocitos T CD8-positivos/metabolismo , Diabetes Mellitus Experimental/inmunología , Ácido Hialurónico/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/prevención & control , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Polimorfismo de Nucleótido Simple/genética , Ratas Endogámicas BB
10.
J Clin Invest ; 117(11): 3507-18, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17965772

RESUMEN

Histamine receptor H1 (H1R) is a susceptibility gene in both experimental autoimmune encephalomyelitis (EAE) and experimental autoimmune orchitis (EAO), 2 classical T cell-mediated models of organ-specific autoimmune disease. Here we showed that expression of H1R in naive CD4+ T cells was required for maximal IFN-gamma production but was dispensable for proliferation. Moreover, H1R signaling at the time of TCR ligation was required for activation of p38 MAPK, a known regulator of IFN-gamma expression. Importantly, selective reexpression of H1R in CD4+ T cells fully complemented both the IFN-gamma production and the EAE susceptibility of H1R-deficient mice. These data suggest that the presence of H1R in CD4+ T cells and its interaction with histamine regulates early TCR signals that lead to Th1 differentiation and autoimmune disease.


Asunto(s)
Interferón gamma/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Histamínicos H1/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Línea Celular , Células Cultivadas , Encefalomielitis Autoinmune Experimental/inmunología , Activación Enzimática , Histamina/inmunología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Orquitis/inmunología , Receptores Histamínicos H1/genética , Transducción de Señal/fisiología , Células TH1/citología , Células TH1/inmunología
11.
Cell Immunol ; 260(2): 119-27, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19914609

RESUMEN

The postnatal maternal environment is known to increase susceptibility to a number of autoimmune diseases. Here we asked whether the postnatal maternal environment could influence autoimmune disease development to day 3 thymectomy (d3tx)-induced autoimmune ovarian disease (AOD) and experimental allergic encephalomyelitis (EAE) in cross-fostered A/J and B6 mice. A/J pups foster-nursed by B6 mothers exhibit an increase in autoimmune disease development while cross-fostering B6 pups on A/J mothers did not alter their susceptibility. The increase in AOD incidence seen in foster-nursed d3tx A/J mice correlated with a decrease in the total number of CD4(+) T cells in the lymph nodes of these animals. Analysis of the cellular composition in the milk revealed that B6 mice shed significantly more maternally derived lymphocytes into their milk compared to A/J mothers. These data suggest that there are maternally derived postnatal factors that influence the development of autoimmune disease in A/J mice.


Asunto(s)
Animales Recién Nacidos/inmunología , Animales Lactantes/inmunología , Enfermedades Autoinmunes/inmunología , Susceptibilidad a Enfermedades/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Quimiocina CXCL1/metabolismo , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Inmunidad Materno-Adquirida/inmunología , Interleucina-13/metabolismo , Interleucina-9/metabolismo , Lactancia/inmunología , Lactancia/metabolismo , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Enfermedades del Ovario/inmunología , Timectomía
12.
Mamm Genome ; 20(3): 162-9, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19205800

RESUMEN

Iddm14 (formerly Iddm4) is a non-MHC-linked genetic locus associated with autoimmune diabetes. Its effects have been well-documented in BB-derived rats in which diabetes is either induced by immunologic perturbation or occurs spontaneously. The role of Iddm14 in non-BB rat strains is unknown. Our goal was to extend the analysis of Iddm14 in new diabetes-susceptible strains and to identify candidate genes in the rat Iddm14 diabetes susceptibility locus that are common to these multiple diabetic strains. To determine if Iddm14 is important in strains other than BB, we first genotyped a (LEW.1WR1 x WF)F2 cohort in which diabetes was induced by perturbation with polyinosinic:polycytidylic acid. We found that Iddm14 is a major determinant of diabetes susceptibility in LEW.1WR1 rats. We then used nucleotide sequencing to establish a strain distribution pattern of polymorphisms (insertions, deletions, and single nucleotide polymorphisms [SNPs]) that predicts susceptibility to diabetes in a panel of inbred and congenic rats. Using the positional information from the congenic strains and the new linkage data, we identified a susceptibility haplotype in the T-cell receptor Vbeta chain (Tcrb-V) locus. This haplotype includes Tcrb-V13, which is identical in five susceptible strains but different in resistant WF and F344 rats. We conclude that Iddm14 is a powerful determinant of both spontaneous and induced autoimmune diabetes in multiple rat strains, and that Tcrb-V13 SNPs constitute a haplotype of gene elements that may be critical for autoimmune diabetes in rats.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Predisposición Genética a la Enfermedad , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Animales , Femenino , Haplotipos , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Ratas , Ratas Endogámicas
13.
Mamm Genome ; 20(11-12): 720-33, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19760323

RESUMEN

MRL mice display unusual healing properties. When MRL ear pinnae are hole punched, the holes close completely without scarring, with regrowth of cartilage and reappearance of both hair follicles and sebaceous glands. Studies using (MRL/lpr x C57BL/6)F(2) and backcross mice first showed that this phenomenon was genetically determined and that multiple loci contributed to this quantitative trait. The lpr mutation itself, however, was not one of them. In the present study we examined the genetic basis of healing in the Large (LG/J) mouse strain, a parent of the MRL mouse and a strain that shows the same healing phenotype. LG/J mice were crossed with Small (SM/J) mice and the F(2) population was scored for healing and their genotypes determined at more than 200 polymorphic markers. As we previously observed for MRL and (MRL x B6)F(2) mice, the wound-healing phenotype was sexually dimorphic, with female mice healing more quickly and more completely than male mice. We found quantitative trait loci (QTLs) on chromosomes (Chrs) 9, 10, 11, and 15. The heal QTLs on Chrs 11 and 15 were linked to differential healing primarily in male animals, whereas QTLs on Chrs 9 and 10 were not sexually dimorphic. A comparison of loci identified in previous crosses with those in the present report using LG/J x SM/J showed that loci on Chrs 9, 11, and 15 colocalized with those seen in previous MRL crosses, whereas the locus on Chr 10 was not seen before and is contributed by SM/J.


Asunto(s)
Sitios de Carácter Cuantitativo , Regeneración/genética , Cicatrización de Heridas/genética , Animales , Mapeo Cromosómico , Cruzamientos Genéticos , Oído/lesiones , Femenino , Haplotipos , Masculino , Ratones , Ratones Endogámicos C57BL , Repeticiones de Microsatélite , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Nucleótido Simple , ARN/química , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Caracteres Sexuales
14.
Am J Pathol ; 173(3): 892-900, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18688020

RESUMEN

Weibel-Palade bodies within endothelial cells are secretory granules known to release von Willebrand Factor (VWF), P-selectin, chemokines, and other stored molecules following histamine exposure. Mice with a disrupted VWF gene (VWFKO) have endothelial cells that are deficient in Weibel-Palade bodies. These mice were used to evaluate the role of VWF and/or Weibel-Palade bodies in Bordetella pertussis toxin-induced hypersensitivity to histamine, a subphenotype of experimental allergic encephalomyelitis, the principal autoimmune model of multiple sclerosis. No significant differences in susceptibility to histamine between wild-type and VWFKO mice were detected after 3 days; however, histamine sensitivity persisted significantly longer in VWFKO mice. Correspondingly, encephalomyelitis onset was earlier, disease was more severe, and blood brain barrier (BBB) permeability was significantly increased in VWFKO mice, as compared with wild-type mice. Moreover, inflammation was selectively increased in the brains, but not spinal cords, of VWFKO mice as compared with wild-type mice. Early increases in BBB permeability in VWFKO mice were not due to increased encephalitogenic T-cell activity since BBB permeability did not differ in adjuvant-treated VWFKO mice as compared with littermates immunized with encephalitogenic peptide plus adjuvant. Taken together, these data indicate that VWF and/or Weibel-Palade bodies negatively regulate BBB permeability changes and autoimmune inflammatory lesion formation within the brain elicited by peripheral inflammatory stimuli.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encéfalo/patología , Permeabilidad Capilar/fisiología , Encefalomielitis Autoinmune Experimental/patología , Factor de von Willebrand/metabolismo , Adyuvantes Inmunológicos/metabolismo , Animales , Barrera Hematoencefálica/patología , Encéfalo/inmunología , Encéfalo/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Histamina/inmunología , Hipersensibilidad/inmunología , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Cuerpos de Weibel-Palade/metabolismo , Factor de von Willebrand/genética
15.
G3 (Bethesda) ; 9(12): 4223-4233, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31645420

RESUMEN

Genetic mapping is a primary tool of genetics in model organisms; however, many quantitative trait loci (QTL) contain tens or hundreds of positional candidate genes. Prioritizing these genes for validation is often ad hoc and biased by previous findings. Here we present a technique for prioritizing positional candidates based on computationally inferred gene function. Our method uses machine learning with functional genomic networks, whose links encode functional associations among genes, to identify network-based signatures of functional association to a trait of interest. We demonstrate the method by functionally ranking positional candidates in a large locus on mouse Chr 6 (45.9 Mb to 127.8 Mb) associated with histamine hypersensitivity (Histh). Histh is characterized by systemic vascular leakage and edema in response to histamine challenge, which can lead to multiple organ failure and death. Although Histh risk is strongly influenced by genetics, little is known about its underlying molecular or genetic causes, due to genetic and physiological complexity of the trait. To dissect this complexity, we ranked genes in the Histh locus by predicting functional association with multiple Histh-related processes. We integrated these predictions with new single nucleotide polymorphism (SNP) association data derived from a survey of 23 inbred mouse strains and congenic mapping data. The top-ranked genes included Cxcl12, Ret, Cacna1c, and Cntn3, all of which had strong functional associations and were proximal to SNPs segregating with Histh. These results demonstrate the power of network-based computational methods to nominate highly plausible quantitative trait genes even in challenging cases involving large QTL and extreme trait complexity.


Asunto(s)
Mapeo Cromosómico , Histamina/genética , Hipersensibilidad/genética , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Animales , Ratones
16.
Diabetes ; 68(7): 1523-1527, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30962219

RESUMEN

Type 1 diabetes (T1D) involves the interaction of multiple gene variants, environmental factors, and immunoregulatory dysfunction. Major T1D genetic risk loci encode HLA-DR and -DQ. Genetic heterogeneity and linkage disequilibrium in the highly polymorphic HLA region confound attempts to identify additional T1D susceptibility loci. To minimize HLA heterogeneity, T1D patients (N = 365) and control subjects (N = 668) homozygous for the HLA-DR3 high-risk haplotype were selected from multiple large T1D studies and examined to identify new T1D susceptibility loci using molecular inversion probe sequencing technology. We report that risk for T1D in HLA-DR3 homozygotes is increased significantly by a previously unreported haplotype of three single nucleotide polymorphisms (SNPs) within the first intron of HLA-DRA1. The homozygous risk haplotype has an odds ratio of 4.65 relative to the protective homozygous haplotype in our sample. Individually, these SNPs reportedly function as "expression quantitative trait loci," modulating HLA-DR and -DQ expression. From our analysis of available data, we conclude that the tri-SNP haplotype within HLA-DRA1 may modulate class II expression, suggesting that increased T1D risk could be attributable to regulated expression of class II genes. These findings could help clarify the role of HLA in T1D susceptibility and improve diabetes risk assessment, particularly in high-risk HLA-DR3 homozygous individuals.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Predisposición Genética a la Enfermedad , Cadenas alfa de HLA-DR/genética , Antígeno HLA-DR3/genética , Polimorfismo de Nucleótido Simple , Alelos , Estudios de Casos y Controles , Femenino , Frecuencia de los Genes , Variación Genética , Genotipo , Homocigoto , Humanos , Intrones , Masculino
17.
Biomaterials ; 197: 244-254, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30669015

RESUMEN

Biological materials are superior to synthetic biomaterials in biocompatibility and active interactions with cells. Here, a new class of biological materials, cell membrane-derived hydrogel scaffolds are reported for harnessing these advantages. To form macroporous scaffolds, vesicles derived from red blood cell membranes (RBCMs) are chemically crosslinked via cryogelation. The RBCM scaffolds with a pore size of around 70 µm are soft and injectable. Highly biocompatible scaffolds are typically made of superhydrophilic polymers and lack the ability to encapsulate and release hydrophobic drugs in a controlled manner. However, hydrophobic molecules can be efficiently encapsulated inside RBCM scaffolds and be sustainedly released. RBCM scaffolds show low neutrophil infiltration after subcutaneous injection in mice, and a significantly higher number of infiltrated macrophages than methacrylate alginate (MA-alginate) scaffolds. According to gene expression and surface markers, these macrophages have an M2-like phenotype, which is anti-inflammatory and immune suppressive. There are also higher percentages of macrophages presenting immunosuppressive PD-L1 in RBCM-scaffolds than in MA-alginate scaffolds. Interestingly, the concentrations of anti-inflammatory cytokine, IL-10 in both types of scaffolds are higher than those in normal organ tissues. This study sheds light on cell membrane-derived hydrogels, which can actively modulate cells in unique ways unavailable to existing hydrogel scaffolds.


Asunto(s)
Materiales Biocompatibles/química , Preparaciones de Acción Retardada/química , Membrana Eritrocítica/química , Andamios del Tejido/química , Animales , Células Cultivadas , Femenino , Macrófagos/citología , Ratones Endogámicos C57BL , Pirenos/administración & dosificación , Ingeniería de Tejidos
18.
Commun Biol ; 2: 398, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31701027

RESUMEN

The systemic capillary leak syndrome (SCLS, Clarkson disease) is a disorder of unknown etiology characterized by recurrent episodes of vascular leakage of proteins and fluids into peripheral tissues, resulting in whole-body edema and hypotensive shock. The pathologic mechanisms and genetic basis for SCLS remain elusive. Here we identify an inbred mouse strain, SJL, which recapitulates cardinal features of SCLS, including susceptibility to histamine- and infection-triggered vascular leak. We named this trait "Histamine hypersensitivity" (Hhs/Hhs) and mapped it to Chromosome 6. Hhs is syntenic to the genomic locus most strongly associated with SCLS in humans (3p25.3), revealing that the predisposition to develop vascular hyperpermeability has a strong genetic component conserved between humans and mice and providing a naturally occurring animal model for SCLS. Genetic analysis of Hhs may reveal orthologous candidate genes that contribute not only to SCLS, but also to normal and dysregulated mechanisms underlying vascular barrier function more generally.


Asunto(s)
Síndrome de Fuga Capilar/genética , Animales , Síndrome de Fuga Capilar/etiología , Síndrome de Fuga Capilar/fisiopatología , Permeabilidad Capilar/genética , Permeabilidad Capilar/fisiología , Mapeo Cromosómico , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Histamina/fisiología , Humanos , Subtipo H3N2 del Virus de la Influenza A , Masculino , Ratones , Ratones Congénicos , Ratones Endogámicos , Infecciones por Orthomyxoviridae/complicaciones , Piel/irrigación sanguínea , Especificidad de la Especie , Sintenía
20.
J Neurosci Res ; 86(12): 2624-40, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18500752

RESUMEN

Pertussis toxin (PTX) is an ancillary adjuvant used to elicit experimental allergic encephalomyelitis (EAE), the principal autoimmune model of multiple sclerosis. One mechanism whereby PTX potentiates EAE is to increase blood-brain barrier (BBB) permeability. To elucidate further the mechanism of action of PTX on the BBB, we investigated the genomic and proteomic responses of isolated mouse brain endothelial cells (MBEC) following intoxication. Among approximately 14,000 mouse genes tracked by cDNA microarray, 34 showed altered expression in response to PTX. More than one-third of these genes have roles in angiogenesis. Accordingly, we show that intoxication of MBEC induces tube formation in vitro and angiogenesis in vivo. The global effect of PTX on signaling protein levels and phosphorylation in MBEC was investigated by using Kinex antibody microarrays. In total, 113 of 372 pan-specific and 58 of 258 phospho-site-specific antibodies revealed changes >or=25% following intoxication. Increased STAT1 Tyr-701 and Ser-727 phosphorylation; reduced phosphorylation of the activating phospho-sites in Erk1, Erk2, and MAPKAPK2; and decreased phosphorylation of arrestin beta1 Ser-412 and Hsp27 Ser-82 were confirmed by Kinetworks multi-immunoblotting. The importance of signal transduction pathways on PTX-induced MBEC tube formation was evaluated pharmacologically. Inhibition of phospholipase C, MEK1, and p38 MAP kinase had little effect, whereas inhibition of cAMP-dependent protein kinase, protein kinase C, and phosphatidylinositol 3-kinase partially blocked tube formation. Taken together, these findings are consistent with the concept that PTX may lead to increased BBB permeability by altering endothelial plasticity and angiogenesis.


Asunto(s)
Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Toxina del Pertussis/farmacología , Animales , Encéfalo/fisiología , Células Cultivadas , Circulación Cerebrovascular/efectos de los fármacos , Circulación Cerebrovascular/genética , Circulación Cerebrovascular/fisiología , Células Endoteliales/fisiología , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Mediadores de Inflamación/farmacología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Microcirculación/efectos de los fármacos , Microcirculación/genética , Microcirculación/fisiología , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA