Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nat Commun ; 8(1): 932, 2017 10 13.
Article in English | MEDLINE | ID: mdl-29030607

ABSTRACT

Inflammatory bowel disease (IBD) involves interaction between host genetic factors and environmental triggers. CCDC88B maps within one IBD risk locus on human chromosome 11q13. Here we show that CCDC88B protein increases in the colon during intestinal injury, concomitant with an influx of CCDC88B+lymphoid and myeloid cells. Loss of Ccdc88b protects against DSS-induced colitis, with fewer pathological lesions and reduced intestinal inflammation in Ccdc88b-deficient mice. In a T cell transfer model of colitis, Ccdc88b mutant CD4+ T cells do not induce colitis in immunocompromised hosts. Expression of human CCDC88B RNA and protein is higher in IBD patient colons than in control colon tissue. In human CD14+ myeloid cells, CCDC88B is regulated by cis-acting variants. In a cohort of patients with Crohn's disease, CCDC88B expression correlates positively with disease risk. These findings suggest that CCDC88B has a critical function in colon inflammation and the pathogenesis of IBD.Hook-related protein family member CCDC88b is encoded by a locus that has been associated with inflammatory bowel disease. Here the authors show that Ccdc88b inactivation in T cells prevents colitis in a transfer model, and detect high colonic levels of CCDC88b in patients with Crohn disease or ulcerative colitis, identifying that expression correlates with disease risk.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Colitis/pathology , Inflammatory Bowel Diseases/pathology , Animals , Colitis/chemically induced , Colitis/metabolism , Colon/metabolism , Colon/pathology , Crohn Disease/metabolism , Crohn Disease/pathology , Dextran Sulfate/toxicity , Gene Expression Regulation , Homeodomain Proteins/genetics , Humans , Inflammatory Bowel Diseases/metabolism , Lipopolysaccharide Receptors/metabolism , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Myeloid Cells/metabolism , Myeloid Cells/pathology , Polymorphism, Single Nucleotide , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
2.
Nat Immunol ; 18(1): 54-63, 2017 01.
Article in English | MEDLINE | ID: mdl-27721430

ABSTRACT

Genes and pathways in which inactivation dampens tissue inflammation present new opportunities for understanding the pathogenesis of common human inflammatory diseases, including inflammatory bowel disease, rheumatoid arthritis and multiple sclerosis. We identified a mutation in the gene encoding the deubiquitination enzyme USP15 (Usp15L749R) that protected mice against both experimental cerebral malaria (ECM) induced by Plasmodium berghei and experimental autoimmune encephalomyelitis (EAE). Combining immunophenotyping and RNA sequencing in brain (ECM) and spinal cord (EAE) revealed that Usp15L749R-associated resistance to neuroinflammation was linked to dampened type I interferon responses in situ. In hematopoietic cells and in resident brain cells, USP15 was coexpressed with, and functionally acted together with the E3 ubiquitin ligase TRIM25 to positively regulate type I interferon responses and to promote pathogenesis during neuroinflammation. The USP15-TRIM25 dyad might be a potential target for intervention in acute or chronic states of neuroinflammation.


Subject(s)
DNA-Binding Proteins/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Malaria, Cerebral/immunology , Neurogenic Inflammation/immunology , Transcription Factors/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , DNA-Binding Proteins/genetics , Encephalomyelitis, Autoimmune, Experimental/drug therapy , HEK293 Cells , Humans , Immunity, Innate , Interferon Type I/metabolism , Malaria, Cerebral/drug therapy , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Molecular Targeted Therapy , Myelin-Oligodendrocyte Glycoprotein/immunology , Neurogenic Inflammation/drug therapy , Peptide Fragments/immunology , Plasmodium berghei/immunology , Transcription Factors/genetics , Ubiquitin-Specific Proteases/genetics
4.
Science ; 346(6214): 1234-8, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25477463

ABSTRACT

Immune and inflammatory responses require leukocytes to migrate within and through the vasculature, a process that is facilitated by their capacity to switch to a polarized morphology with an asymmetric distribution of receptors. We report that neutrophil polarization within activated venules served to organize a protruding domain that engaged activated platelets present in the bloodstream. The selectin ligand PSGL-1 transduced signals emanating from these interactions, resulting in the redistribution of receptors that drive neutrophil migration. Consequently, neutrophils unable to polarize or to transduce signals through PSGL-1 displayed aberrant crawling, and blockade of this domain protected mice against thromboinflammatory injury. These results reveal that recruited neutrophils scan for activated platelets, and they suggest that the neutrophils' bipolarity allows the integration of signals present at both the endothelium and the circulation before inflammation proceeds.


Subject(s)
Blood Platelets/immunology , Inflammation/immunology , Neutrophils/immunology , Platelet Activation , Thrombosis/immunology , Animals , Blood Circulation , Cell Movement , Cell Polarity , Endothelium, Vascular/immunology , Inflammation/blood , Male , Membrane Glycoproteins , Mice , Mice, Inbred C57BL , Signal Transduction , Venules/immunology
5.
PLoS Pathog ; 10(12): e1004511, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25473962

ABSTRACT

Natural Killer (NK) cells contribute to the control of viral infection by directly killing target cells and mediating cytokine release. In C57BL/6 mice, the Ly49H activating NK cell receptor plays a key role in early resistance to mouse cytomegalovirus (MCMV) infection through specific recognition of the MCMV-encoded MHC class I-like molecule m157 expressed on infected cells. Here we show that transgenic expression of Ly49H failed to provide protection against MCMV infection in the naturally susceptible A/J mouse strain. Characterization of Ly49H(+) NK cells from Ly49h-A transgenic animals showed that they were able to mount a robust cytotoxic response and proliferate to high numbers during the course of infection. However, compared to NK cells from C57BL/6 mice, we observed an intrinsic defect in their ability to produce IFNγ when challenged by either m157-expressing target cells, exogenous cytokines or chemical stimulants. This effect was limited to NK cells as T cells from C57BL/6 and Ly49h-A mice produced comparable cytokine levels. Using a panel of recombinant congenic strains derived from A/J and C57BL/6 progenitors, we mapped the genetic basis of defective IFNγ production to a single 6.6 Mb genetic interval overlapping the Ifng gene on chromosome 10. Inspection of the genetic interval failed to reveal molecular differences between A/J and several mouse strains showing normal IFNγ production. The chromosome 10 locus is independent of MAPK signalling or decreased mRNA stability and linked to MCMV susceptibility. This study highlights the existence of a previously uncovered NK cell-specific cis-regulatory mechanism of Ifnγ transcript expression potentially relevant to NK cell function in health and disease.


Subject(s)
Cytomegalovirus Infections/genetics , Cytomegalovirus , Genetic Loci , Genetic Predisposition to Disease , Interferon-gamma/genetics , Animals , Chromosomes, Mammalian , Cytomegalovirus Infections/immunology , Gene Expression Regulation, Viral/genetics , Gene Expression Regulation, Viral/immunology , Interferon-gamma/immunology , Mice , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily A , RNA Stability/genetics , RNA Stability/immunology , Viral Proteins/genetics , Viral Proteins/immunology
6.
J Immunol ; 193(3): 1290-300, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24973457

ABSTRACT

In the immunocompromised host, invasive infection with the fungal pathogen Candida albicans is associated with high morbidity and mortality. Sporadic cases in otherwise normal individuals are rare, and they are thought to be associated with genetic predisposition. Using a mouse model of systemic infection with C. albicans, we identified the SM/J mouse strain as unusually susceptible to infection. Genetic linkage studies in informative [C57BL/6JxSM/J]F2 mice identified a major locus on distal chromosome 15, given the appellation Carg5, that regulates C. albicans replication in SM/J mice. Cellular and molecular immunophenotyping experiments, as well as functional studies in purified cell populations from SM/J and C57BL/6J, and in [C57BL/6JxSM/J]F2 mice fixed for homozygous or heterozygous Carg5 alleles, indicate that Carg5-regulated susceptibility in SM/J is associated with a complex defect in the myeloid compartment of these mice. SM/J neutrophils express lower levels of Ly6G, and importantly, they show significantly reduced production of reactive oxygen species in response to stimulation with fMLF and PMA. Likewise, CD11b(+)Ly6G(-)Ly6C(hi) inflammatory monocytes were present at lower levels in the blood of infected SM/J, recruited less efficiently at the site of infection, and displayed blunted oxidative burst. Studies in F2 mice establish strong correlations between Carg5 alleles, Ly6G expression, production of serum CCL2 (MCP-1), and susceptibility to C. albicans. Genomic DNA sequencing of chromatin immunoprecipitated for myeloid proinflammatory transcription factors IRF1, IRF8, STAT1 and NF-κB, as well as RNA sequencing, were used to develop a "myeloid inflammatory score" and systematically analyze and prioritize potential candidate genes in the Carg5 interval.


Subject(s)
Candidiasis/genetics , Candidiasis/immunology , Genetic Predisposition to Disease , Animals , Antigens, Ly/biosynthesis , Antigens, Ly/genetics , Candidiasis/microbiology , Chemokine CCL2/biosynthesis , Chemokine CCL2/blood , Disease Models, Animal , Disease Susceptibility/immunology , Disease Susceptibility/microbiology , Female , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Immunophenotyping , Inflammation/genetics , Inflammation/immunology , Inflammation/microbiology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred A , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/microbiology , Monocytes/pathology , Neutrophils/immunology , Neutrophils/microbiology , Neutrophils/pathology , Primary Cell Culture , Repressor Proteins/genetics , Species Specificity , Spores, Fungal/genetics , Spores, Fungal/growth & development , Spores, Fungal/immunology
7.
PLoS Pathog ; 9(7): e1003491, 2013.
Article in English | MEDLINE | ID: mdl-23853600

ABSTRACT

Interferon Regulatory Factor 8 (IRF8) is required for development, maturation and expression of anti-microbial defenses of myeloid cells. BXH2 mice harbor a severely hypomorphic allele at Irf8 (Irf8(R294C)) that causes susceptibility to infection with intracellular pathogens including Mycobacterium tuberculosis. We report that BXH2 are completely resistant to the development of cerebral malaria (ECM) following Plasmodium berghei ANKA infection. Comparative transcriptional profiling of brain RNA as well as chromatin immunoprecipitation and high-throughput sequencing (ChIP-seq) was used to identify IRF8-regulated genes whose expression is associated with pathological acute neuroinflammation. Genes increased by infection were strongly enriched for IRF8 binding sites, suggesting that IRF8 acts as a transcriptional activator in inflammatory programs. These lists were enriched for myeloid-specific pathways, including interferon responses, antigen presentation and Th1 polarizing cytokines. We show that inactivation of several of these downstream target genes (including the Irf8 transcription partner Irf1) confers protection against ECM. ECM-resistance in Irf8 and Irf1 mutants is associated with impaired myeloid and lymphoid cells function, including production of IL12p40 and IFNγ. We note strong overlap between genes bound and regulated by IRF8 during ECM and genes regulated in the lungs of M. tuberculosis infected mice. This IRF8-dependent network contains several genes recently identified as risk factors in acute and chronic human inflammatory conditions. We report a common core of IRF8-bound genes forming a critical inflammatory host-response network.


Subject(s)
Brain/immunology , Gene Expression Regulation , Immunity, Innate , Interferon Regulatory Factors/metabolism , Malaria, Cerebral/immunology , Nerve Tissue Proteins/metabolism , Plasmodium berghei/immunology , Amino Acid Substitution , Animals , Binding Sites , Brain/metabolism , Brain/parasitology , Cells, Cultured , Cytokines/biosynthesis , Cytokines/blood , Gene Expression Profiling , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factors/chemistry , Interferon Regulatory Factors/genetics , Malaria, Cerebral/blood , Malaria, Cerebral/metabolism , Malaria, Cerebral/parasitology , Mice , Mice, Knockout , Mice, Mutant Strains , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurons/immunology , Neurons/metabolism , Neurons/parasitology , Specific Pathogen-Free Organisms , Spleen/immunology , Spleen/metabolism , Spleen/pathology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/parasitology
8.
Science ; 337(6102): 1684-8, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22859821

ABSTRACT

ISG15 is an interferon (IFN)-α/ß-inducible, ubiquitin-like intracellular protein. Its conjugation to various proteins (ISGylation) contributes to antiviral immunity in mice. Here, we describe human patients with inherited ISG15 deficiency and mycobacterial, but not viral, diseases. The lack of intracellular ISG15 production and protein ISGylation was not associated with cellular susceptibility to any viruses that we tested, consistent with the lack of viral diseases in these patients. By contrast, the lack of mycobacterium-induced ISG15 secretion by leukocytes-granulocyte, in particular-reduced the production of IFN-γ by lymphocytes, including natural killer cells, probably accounting for the enhanced susceptibility to mycobacterial disease. This experiment of nature shows that human ISGylation is largely redundant for antiviral immunity, but that ISG15 plays an essential role as an IFN-γ-inducing secreted molecule for optimal antimycobacterial immunity.


Subject(s)
Cytokines/immunology , Interferon-gamma/immunology , Mycobacterium Infections/immunology , Ubiquitins/immunology , Virus Diseases/immunology , Animals , Antibodies, Viral/blood , Cytokines/genetics , Female , Granulocytes/immunology , Humans , Immunity , Interleukin-12/immunology , Killer Cells, Natural/immunology , Male , Mice , Mycobacterium Infections/blood , Mycobacterium Infections/genetics , Pedigree , T-Lymphocytes/immunology , Ubiquitins/genetics , Virus Diseases/blood
9.
PLoS Negl Trop Dis ; 5(12): e1435, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22206032

ABSTRACT

Human neurocysticercosis (NC) caused by Taenia solium is a parasitic disease of the central nervous system that is endemic in many developing countries. In this study, a genetic approach using the murine intraperitoneal cysticercosis caused by the related cestode Taenia crassiceps was employed to identify host factors that regulate the establishment and proliferation of the parasite. A/J mice are permissive to T. crassiceps infection while C57BL/6J mice (B6) are comparatively restrictive, with a 10-fold difference in numbers of peritoneal cysticerci recovered 30 days after infection. The genetic basis of this inter-strain difference was explored using 34 AcB/BcA recombinant congenic strains derived from A/J and B6 progenitors, that were phenotyped for T. crassiceps replication. In agreement with their genetic background, most AcB strains (A/J-derived) were found to be permissive to infection while most BcA strains (B6-derived) were restrictive with the exception of a few discordant strains, together suggesting a possible simple genetic control. Initial haplotype association mapping using >1200 informative SNPs pointed to linkages on chromosomes 2 (proximal) and 6 as controlling parasite replication in the AcB/BcA panel. Additional linkage analysis by genome scan in informative [AcB55xDBA/2]F1 and F2 mice (derived from the discordant AcB55 strain), confirmed the effect of chromosome 2 on parasite replication, and further delineated a major locus (LOD = 4.76, p<0.01; peak marker D2Mit295, 29.7 Mb) that we designate Tccr1 (T. crassiceps cysticercosis restrictive locus 1). Resistance alleles at Tccr1 are derived from AcB55 and are inherited in a dominant fashion. Scrutiny of the minimal genetic interval reveals overlap of Tccr1 with other host resistance loci mapped to this region, most notably the defective Hc/C5 allele which segregates both in the AcB/BcA set and in the AcB55xDBA/2 cross. These results strongly suggest that the complement component 5 (C5) plays a critical role in early protective inflammatory response to infection with T. crassiceps.


Subject(s)
Complement C5/genetics , Complement C5/immunology , Cysticercosis/genetics , Cysticercosis/immunology , Taenia/immunology , Animals , Crosses, Genetic , Disease Models, Animal , Female , Genetic Association Studies , Haplotypes , Mice , Mice, Inbred A , Mice, Inbred C57BL , Polymorphism, Single Nucleotide
10.
PLoS One ; 6(4): e18957, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21533108

ABSTRACT

Candida albicans is an opportunistic pathogen that causes acute disseminated infections in immunocompromised hosts, representing an important cause of morbidity and mortality in these patients. To study the genetic control of susceptibility to disseminated C. albicans in mice, we phenotyped a group of 23 phylogenetically distant inbred strains for susceptibility to infection as measured by extent of fungal replication in the kidney 48 hours following infection. Susceptibility was strongly associated with the loss-of-function mutant complement component 5 (C5/Hc) allele, which is known to be inherited by approximately 40% of inbred strains. Our survey identified 2 discordant strains, AKR/J (C5-deficient, resistant) and SM/J (C5-sufficient, susceptible), suggesting that additional genetic effects may control response to systemic candidiasis in these strains. Haplotype association mapping in the 23 strains using high density SNP maps revealed several putative loci regulating the extent of C. albicans replication, amongst which the most significant were C5 (P value = 2.43×10(-11)) and a novel effect on distal chromosome 11 (P value = 7.63×10(-9)). Compared to other C5-deficient strains, infected AKR/J strain displays a reduced fungal burden in the brain, heart and kidney, and increased survival, concomitant with uniquely high levels of serum IFNγ. C5-independent genetic effects were further investigated by linkage analysis in an [A/JxAKR/J]F2 cross (n = 158) where the mutant Hc allele is fixed. These studies identified a chromosome 11 locus (Carg4, Candida albicans resistance gene 4; LOD = 4.59), and a chromosome 8 locus (Carg3; LOD = 3.95), both initially detected by haplotype association mapping. Alleles at both loci were inherited in a co-dominant manner. Our results verify the important effect of C5-deficiency in inbred mouse strains, and further identify two novel loci, Carg3 and Carg4, which regulate resistance to C. albicans infection in a C5-independent manner.


Subject(s)
Candida albicans/isolation & purification , Candidiasis/genetics , Genetic Predisposition to Disease , Animals , Candida albicans/growth & development , Candidiasis/microbiology , Chromosome Mapping , Complement C5/genetics , Haplotypes , Lod Score , Mice , Mice, Inbred Strains , Phenotype , Polymorphism, Single Nucleotide
11.
Exp Parasitol ; 125(4): 315-24, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20219464

ABSTRACT

In mice, loss of pantetheinase activity causes susceptibility to infection with Plasmodium chabaudi AS. Treatment of mice with the pantetheinase metabolite cysteamine reduces blood-stage replication of P. chabaudi and significantly increases survival. Similarly, a short exposure of Plasmodium to cysteamine ex vivo is sufficient to suppress parasite infectivity in vivo. This effect of cysteamine is specific and not observed with a related thiol (dimercaptosuccinic acid) or with the pantethine precursor of cysteamine. Also, cysteamine does not protect against infection with the parasite Trypanosoma cruzi or the fungal pathogen Candida albicans, suggesting cysteamine acts directly against the parasite and does not modulate host inflammatory response. Cysteamine exposure also blocks replication of P. falciparum in vitro; moreover, these treated parasites show higher levels of intact hemoglobin. This study highlights the in vivo action of cysteamine against Plasmodium and provides further evidence for the involvement of pantetheinase in host response to this infection.


Subject(s)
Antimalarials/pharmacology , Cysteamine/pharmacology , Malaria/drug therapy , Plasmodium chabaudi/drug effects , Plasmodium falciparum/drug effects , Amidohydrolases/metabolism , Animals , Antimalarials/therapeutic use , Candidiasis/drug therapy , Chagas Disease/drug therapy , Chloroquine/pharmacology , Cysteamine/therapeutic use , Cytokines/blood , Cytokines/drug effects , Dose-Response Relationship, Drug , Erythrocytes/drug effects , Erythrocytes/parasitology , Female , GPI-Linked Proteins , Hemoglobins/metabolism , Humans , Malaria/parasitology , Male , Mice , Mice, Inbred C57BL , Parasitemia/drug therapy , Parasitemia/parasitology , Plasmodium falciparum/metabolism , Trypanosoma cruzi/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...