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










Publication year range
1.
Commun Biol ; 7(1): 77, 2024 01 10.
Article in English | MEDLINE | ID: mdl-38200184

ABSTRACT

CCDC88B is a risk factor for several chronic inflammatory diseases in humans and its inactivation causes a migratory defect in DCs in mice. CCDC88B belongs to a family of cytoskeleton-associated scaffold proteins that feature protein:protein interaction domains. Here, we identified the Rho/Rac Guanine Nucleotide Exchange Factor 2 (ARHGEF2) and the RAS Protein Activator Like 3 (RASAL3) as CCDC88B physical and functional interactors. Mice defective in Arhgef2 or Rasal3 show dampened neuroinflammation, and display altered cellular response and susceptibility to colitis; ARHGEF2 maps to a human Chromosome 1 locus associated with susceptibility to IBD. Arhgef2 and Rasal3 mutant DCs show altered migration and motility in vitro, causing either reduced (Arhgef2) or enhanced (Rasal3) migratory properties. The CCDC88B/RASAL3/ARHGEF2 complex appears to regulate DCs migration by modulating activation of RHOA, with ARHGEF2 and RASAL3 acting in opposite regulatory fashions, providing a molecular mechanism for the involvement of these proteins in DCs immune functions.


Subject(s)
Colitis , Neuroinflammatory Diseases , Animals , Humans , Mice , Cell Physiological Phenomena , Colitis/genetics , Cytoskeleton , Dendritic Cells , Rho Guanine Nucleotide Exchange Factors/genetics
2.
Transfus Med Rev ; 37(3): 150748, 2023 07.
Article in English | MEDLINE | ID: mdl-37827586

ABSTRACT

Biphosphoglycerate mutase (BPGM) is a tri-functional enzyme expressed exclusively in erythroid cells and tissues that is responsible for the production of 2,3-biphosphoglycerate (2,3-BPG) through the Rapoport-Luebering shunt. The 2,3-BPG is required for efficient glycolysis and ATP production under anaerobic conditions, but is also a critical allosteric regulator of hemoglobin (Hb), acting to regulate oxygen release in peripheral tissues. In humans, BPGM deficiency is very rare, and is associated with reduced levels of erythrocytic 2,3-BPG and ATP, left shifted Hb-O2 dissociation curve, low P50, elevated Hb and constitutive erythrocytosis. BPGM deficiency in mice recapitulates the erythroid defects seen in human patients. A recent report has shown that BPGM deficiency in mice affords striking protection against both severe malaria anemia and cerebral malaria. These findings are reminiscent of studies of another erythrocyte specific glycolytic enzyme, Pyruvate Kinase (PKLR), which mutational inactivation protects humans and mice against malaria through impairment of glycolysis and ATP production in erythrocytes. BPGM, and PKLR join glucose-6-phosphate dehydrogenase (G6PD) and other erythrocyte variants as modulating response to malaria. Recent studies reviewed suggest glycolysis in general, and BPGM in particular, as a novel pharmacological target for therapeutic intervention in malaria.


Subject(s)
Intramolecular Transferases , Malaria , Humans , Mice , Animals , Erythrocytes , Hemoglobins , Oxygen , Malaria/drug therapy , Adenosine Triphosphate
3.
Sci Rep ; 11(1): 15073, 2021 07 23.
Article in English | MEDLINE | ID: mdl-34302001

ABSTRACT

The estrogen-related receptor alpha (ERRα) is a primary regulator of mitochondrial energy metabolism, function and dynamics, and has been implicated in autophagy and immune regulation. ERRα is abundantly expressed in the intestine and in cells of the immune system. However, its role in inflammatory bowel disease (IBD) remains unknown. Here, we report a protective role of ERRα in the intestine. We found that mice deficient in ERRα were susceptible to experimental colitis, exhibiting increased colon inflammation and tissue damage. This phenotype was mediated by impaired compensatory proliferation of intestinal epithelial cells (IEC) following injury, enhanced IEC apoptosis and necrosis and reduced mucus-producing goblet cell counts. Longitudinal analysis of the microbiota demonstrated that loss of ERRα lead to a reduction in microbiome α-diversity and depletion of healthy gut bacterial constituents. Mechanistically, ERRα mediated its protective effects by acting within the radio-resistant compartment of the intestine. It promoted disease tolerance through transcriptional control of key genes involved in intestinal tissue homeostasis and repair. These findings provide new insights on the role of ERRα in the gut and extends our current knowledge of nuclear receptors implicated in IBD.


Subject(s)
Colitis/genetics , Energy Metabolism/genetics , Inflammatory Bowel Diseases/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Receptors, Estrogen/genetics , Animals , Apoptosis/genetics , Cell Proliferation/genetics , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Colon/metabolism , Colon/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Homeostasis/genetics , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/pathology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Microbiota/genetics , Necrosis/genetics , Necrosis/metabolism , Necrosis/pathology , ERRalpha Estrogen-Related Receptor
4.
Genes Immun ; 22(1): 12-23, 2021 05.
Article in English | MEDLINE | ID: mdl-33824497

ABSTRACT

The covalent post-translational modification of proteins by ubiquitination not only influences protein stability and half-life, but also several aspects of protein function including enzymatic activity, sub-cellular localization, and interactions with binding partners. Protein ubiquitination status is determined by the action of large families of ubiquitin ligases and deubiquitinases, whose combined activities regulate many physiological and cellular pathways. The Ubiquitin Specific Protease (USP) family is one of 8 subfamilies of deubiquitinating enzymes composed of more than 50 members. Recent studies have shown that USP15 plays a critical role in regulating many aspects of immune and inflammatory function of leukocytes in response to a broad range of infectious and autoimmune insults and following tissue damage. USP15 regulated pathways reviewed herein include TLR signaling, RIG-I signaling, NF-kB, and IRF3/IRF7-dependent transcription for production of pro-inflammatory cytokines and type I interferons. In addition, USP15 has been found to regulate pathways implicated in tumor onset and progression such as p53, and TGF-ß signaling, but also influences the leukocytes-determined immune and inflammatory microenvironment of tumors to affect progression and outcome. Hereby reviewed are recent studies of USP15 in model cell lines in vitro, and in mutant mice in vivo with reference to available human clinical datasets.


Subject(s)
Interferon Type I , Ubiquitin-Specific Proteases , Animals , Mice , NF-kappa B/metabolism , Signal Transduction , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , Ubiquitination
5.
Cell Rep ; 32(12): 108170, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32966787

ABSTRACT

The replication cycle and pathogenesis of the Plasmodium malarial parasite involves rapid expansion in red blood cells (RBCs), and variants of certain RBC-specific proteins protect against malaria in humans. In RBCs, bisphosphoglycerate mutase (BPGM) acts as a key allosteric regulator of hemoglobin/oxyhemoglobin. We demonstrate here that a loss-of-function mutation in the murine Bpgm (BpgmL166P) gene confers protection against both Plasmodium-induced cerebral malaria and blood-stage malaria. The malaria protection seen in BpgmL166P mutant mice is associated with reduced blood parasitemia levels, milder clinical symptoms, and increased survival. The protective effect of BpgmL166P involves a dual mechanism that enhances the host's stress erythroid response to Plasmodium-driven RBC loss and simultaneously alters the intracellular milieu of the RBCs, including increased oxyhemoglobin and reduced energy metabolism, reducing Plasmodium maturation, and replication. Overall, our study highlights the importance of BPGM as a regulator of hemoglobin/oxyhemoglobin in malaria pathogenesis and suggests a new potential malaria therapeutic target.


Subject(s)
Anemia/etiology , Anemia/prevention & control , Bisphosphoglycerate Mutase/deficiency , Malaria, Cerebral/enzymology , Malaria, Cerebral/prevention & control , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , Bisphosphoglycerate Mutase/chemistry , Bisphosphoglycerate Mutase/genetics , Bisphosphoglycerate Mutase/metabolism , Enzyme Stability , Erythrocytes/enzymology , Erythrocytes/parasitology , Erythropoiesis , Extracellular Matrix/metabolism , Female , HEK293 Cells , Humans , Malaria, Cerebral/complications , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation/genetics , Parasites/growth & development , Plasmodium/growth & development , Polycythemia
6.
J Leukoc Biol ; 108(6): 1787-1802, 2020 12.
Article in English | MEDLINE | ID: mdl-32480428

ABSTRACT

The Coiled Coil Domain Containing Protein 88B (CCDC88B) gene is associated with susceptibility to several inflammatory diseases in humans and its inactivation in mice protects against acute neuroinflammation and models of intestinal colitis. We report that mice lacking functional CCDC88B (Ccdc88bMut ) are defective in several dendritic cells (DCs)-dependent inflammatory and immune reactions in vivo. In these mice, an inflammatory stimulus (LPS) fails to induce the recruitment of DCs into the draining lymph nodes (LNs). In addition, OVA-pulsed Ccdc88bMut DCs injected in the footpad do not induce recruitment and activation of antigen-specific CD4+ and CD8+ T cells in their draining LN. Experiments in vitro indicate that this defect is independent of the ability of mutant DCs to capture and present peptide antigen to T cells. Rather, kinetic analyses in vivo of wild-type and Ccdc88bMut DCs indicate a reduced migration capacity in the absence of the CCDC88B protein expression. Moreover, using time-lapse light microscopy imaging, we show that Ccdc88bMut DCs have an intrinsic motility defect. Furthermore, in vivo studies reveal that these reduced migratory properties lead to dampened contact hypersensitivity reactions in Ccdc88b mutant mice. These findings establish a critical role of CCDC88B in regulating movement and migration of DCs. Thus, regulatory variants impacting Ccdc88b expression in myeloid cells may cause variable degrees of DC-dependent inflammatory response in situ, providing a rationale for the genetic association of CCDC88B with several inflammatory and autoimmune diseases in humans.


Subject(s)
Antigen Presentation , Carrier Proteins/immunology , Cell Movement/immunology , Dendritic Cells/immunology , Gene Expression Regulation/immunology , Animals , Carrier Proteins/genetics , Cell Movement/genetics , Dendritic Cells/cytology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Transgenic
7.
Sci Rep ; 9(1): 18897, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31827213

ABSTRACT

The mechanisms linking chronic inflammation of the gut (IBD) and increased colorectal cancer susceptibility are poorly understood. IBD risk is influenced by genetic factors, including the IBD5 locus (human 5q31), that harbors the IRF1 gene. A cause-to-effect relationship between chronic inflammation and colorectal cancer, and a possible role of IRF1 were studied in Irf1-/- mice in a model of colitis-associated colorectal cancer (CA-CRC) induced by azoxymethane and dextran sulfate. Loss of Irf1 causes hyper-susceptibility to CA-CRC, with early onset and increased number of tumors leading to rapid lethality. Transcript profiling (RNA-seq) and immunostaining of colons shows heightened inflammation and enhanced enterocyte proliferation in Irf1-/- mutants, prior to appearance of tumors. Considerable infiltration of leukocytes is seen in Irf1-/- colons at this early stage, and is composed primarily of proinflammatory Gr1+ Cd11b+ myeloid cells and other granulocytes, as well as CD4+ lymphoid cells. Differential susceptibility to CA-CRC of Irf1-/- vs. B6 controls is fully transferable through hematopoietic cells as observed in bone marrow chimera studies. Transcript signatures seen in Irf1-/- mice in response to AOM/DSS are enriched in clinical specimens from patients with IBD and with colorectal cancer. In addition, IRF1 expression in the colon is significantly decreased in late stage colorectal cancer (stages 3, 4) and is associated with poorer prognosis. This suggests that partial or complete loss of IRF1 expression alters the type, number, and function of immune cells in situ during chronic inflammation, possibly via the creation of a tumor-promoting environment.


Subject(s)
Colitis/metabolism , Colorectal Neoplasms/metabolism , Interferon Regulatory Factor-1/metabolism , Animals , Colitis/complications , Colitis/genetics , Colitis/pathology , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Models, Animal , Genetic Predisposition to Disease , Interferon Regulatory Factor-1/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Knockout
8.
Structure ; 27(4): 590-605.e5, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30713027

ABSTRACT

The multi-domain deubiquitinase USP15 regulates diverse eukaryotic processes and has been implicated in numerous diseases. We developed ubiquitin variants (UbVs) that targeted either the catalytic domain or each of three adaptor domains in USP15, including the N-terminal DUSP domain. We also designed a linear dimer (diUbV), which targeted the DUSP and catalytic domains, and exhibited enhanced specificity and more potent inhibition of catalytic activity than either UbV alone. In cells, the UbVs inhibited the deubiquitination of two USP15 substrates, SMURF2 and TRIM25, and the diUbV inhibited the effects of USP15 on the transforming growth factor ß pathway. Structural analyses revealed that three distinct UbVs bound to the catalytic domain and locked the active site in a closed, inactive conformation, and one UbV formed an unusual strand-swapped dimer and bound two DUSP domains simultaneously. These inhibitors will enable the study of USP15 function in oncology, neurology, immunology, and inflammation.


Subject(s)
Transcription Factors/chemistry , Transforming Growth Factor beta1/chemistry , Tripartite Motif Proteins/chemistry , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Specific Proteases/chemistry , Ubiquitin/chemistry , Amino Acid Sequence , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , Ubiquitination
9.
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
10.
Immunology ; 151(1): 110-121, 2017 05.
Article in English | MEDLINE | ID: mdl-28066899

ABSTRACT

Several previous studies outlined the importance of the histone H2A deubiquitinase MYSM1 in the regulation of stem cell quiescence and haematopoiesis. In this study we investigated the role of MYSM1 in T-cell development. Using mouse models that allow conditional Mysm1 ablation at late stages of thymic development, we found that MYSM1 is intricately involved in the maintenance, activation and survival of CD8+ T cells. Mysm1 ablation resulted in a twofold reduction in CD8+ T-cell numbers, and also led to a hyperactivated CD8+ T-cell state accompanied by impaired proliferation and increased pro-inflammatory cytokine production after ex vivo stimulation. These phenotypes coincided with an increased apoptosis and preferential up-regulation of p53 tumour suppressor protein in CD8+ T cells. Lastly, we examined a model of experimental cerebral malaria, in which pathology is critically dependent on CD8+ T cells. In the mice conditionally deleted for Mysm1 in the T-cell compartment, CD8+ T-cell numbers remained reduced following infection, both in the periphery and in the brain, and the mice displayed improved survival after parasite challenge. Collectively, our data identify MYSM1 as a novel factor for CD8+ T cells in the immune system, increasing our understanding of the role of histone H2A deubiquitinases in cytotoxic T-cell biology.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Endopeptidases/metabolism , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Cytokines/metabolism , Cytotoxicity, Immunologic/genetics , Endopeptidases/genetics , Inflammation Mediators/metabolism , Lymphocyte Activation/genetics , Malaria, Cerebral/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Trans-Activators , Tumor Suppressor Protein p53/genetics , Ubiquitin-Specific Proteases
11.
Annu Rev Immunol ; 35: 1-30, 2017 04 26.
Article in English | MEDLINE | ID: mdl-27912315

ABSTRACT

Genome technologies have defined a complex genetic architecture in major infectious, inflammatory, and autoimmune disorders. High density marker arrays and Immunochips have powered genome-wide association studies (GWAS) that have mapped nearly 450 genetic risk loci in 22 major inflammatory diseases, including a core of common genes that play a central role in pathological inflammation. Whole-exome and whole-genome sequencing have identified more than 265 genes in which mutations cause primary immunodeficiencies and rare forms of severe inflammatory bowel disease. Combined analysis of inflammatory disease GWAS and primary immunodeficiencies point to shared proteins and pathways that are required for immune cell development and protection against infections and are also associated with pathological inflammation. Finally, sequencing of chromatin immunoprecipitates containing specific transcription factors, with parallel RNA sequencing, has charted epigenetic regulation of gene expression by proinflammatory transcription factors in immune cells, providing complementary information to characterize morbid genes at infectious and inflammatory disease loci.


Subject(s)
Autoimmune Diseases/genetics , Immunologic Deficiency Syndromes/genetics , Infections/genetics , Inflammation/genetics , Vaccines/immunology , Animals , Epigenesis, Genetic , Exome/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing , Humans , Immunity/genetics , Infections/immunology , Risk
12.
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
14.
Trends Immunol ; 37(2): 126-140, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26791050

ABSTRACT

Recent advances in genome analysis have provided important insights into the genetic architecture of infectious and inflammatory diseases. The combined analysis of loci detected by genome-wide association studies (GWAS) in 22 inflammatory diseases has revealed a shared genetic core and associated biochemical pathways that play a central role in pathological inflammation. Parallel whole-exome sequencing studies have identified 265 genes mutated in primary immunodeficiencies (PID). Here, we examine the overlap between these two data sets, and find that it consists of genes essential for protection against infections and in which persistent activation causes pathological inflammation. Based on this intersection, we propose that, although strong or inactivating mutations (rare variants) in these genes may cause severe disease (PIDs), their more subtle modulation potentially by common regulatory/coding variants may contribute to chronic inflammation.


Subject(s)
Autoimmune Diseases/genetics , Genetic Loci/immunology , Immunologic Deficiency Syndromes/genetics , Animals , Exome/immunology , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Mutation/genetics , Risk
15.
Infect Immun ; 83(2): 759-68, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25452553

ABSTRACT

We identify an N-ethyl-N-nitrosourea (ENU)-induced I23N mutation in the THEMIS protein that causes protection against experimental cerebral malaria (ECM) caused by infection with Plasmodium berghei ANKA. Themis(I23N) homozygous mice show reduced CD4(+) and CD8(+) T lymphocyte numbers. ECM resistance in P. berghei ANKA-infected Themis(I23N) mice is associated with decreased cerebral cellular infiltration, retention of blood-brain barrier integrity, and reduced proinflammatory cytokine production. THEMIS(I23N) protein expression is absent from mutant mice, concurrent with the decreased THEMIS(I23N) stability observed in vitro. Biochemical studies in vitro and functional complementation in vivo in Themis(I23N/+):Lck(-/+) doubly heterozygous mice demonstrate that functional coupling of THEMIS to LCK tyrosine kinase is required for ECM pathogenesis. Damping of proinflammatory responses in Themis(I23N) mice causes susceptibility to pulmonary tuberculosis. Thus, THEMIS is required for the development and ultimately the function of proinflammatory T cells. Themis(I23N) mice can be used to study the newly discovered association of THEMIS (6p22.33) with inflammatory bowel disease and multiple sclerosis.


Subject(s)
Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Proteins/genetics , Tuberculosis, Pulmonary/immunology , Animals , Blood-Brain Barrier , Brain/pathology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Celiac Disease/genetics , Ethylnitrosourea , Gene Expression , Inflammation/immunology , Intercellular Signaling Peptides and Proteins , Malaria, Cerebral/parasitology , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Parasitemia/pathology , Proteins/immunology , Tuberculosis, Pulmonary/microbiology
16.
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
17.
J Exp Med ; 211(13): 2519-35, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25403443

ABSTRACT

We used a genome-wide screen in mutagenized mice to identify genes which inactivation protects against lethal neuroinflammation during experimental cerebral malaria (ECM). We identified an ECM-protective mutation in coiled-coil domain containing protein 88b (Ccdc88b), a poorly annotated gene that is found expressed specifically in spleen, bone marrow, lymph nodes, and thymus. The CCDC88B protein is abundantly expressed in immune cells, including both CD4(+) and CD8(+) T lymphocytes, and in myeloid cells, and loss of CCDC88B protein expression has pleiotropic effects on T lymphocyte functions, including impaired maturation in vivo, significantly reduced activation, reduced cell division as well as impaired cytokine production (IFN-γ and TNF) in response to T cell receptor engagement, or to nonspecific stimuli in vitro, and during the course of P. berghei infection in vivo. This identifies CCDC88B as a novel and important regulator of T cell function. The human CCDC88B gene maps to the 11q13 locus that is associated with susceptibility to several inflammatory and auto-immune disorders. Our findings strongly suggest that CCDC88B is the morbid gene underlying the pleiotropic effect of the 11q13 locus on inflammation.


Subject(s)
Carrier Proteins/genetics , Cell Differentiation , Inflammation/immunology , Inflammation/pathology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Base Sequence , Carrier Proteins/metabolism , Chromosomes, Human, Pair 11/genetics , Disease Resistance/immunology , Ethylnitrosourea , Female , Gene Expression Regulation , Genetic Association Studies , Hematopoietic System/metabolism , Humans , Lymphocyte Activation/immunology , Malaria, Cerebral/genetics , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Malaria, Cerebral/prevention & control , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Sequence Data , Mutation/genetics , Myeloid Cells/metabolism , Organ Specificity/genetics , Plasmodium berghei , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/metabolism
19.
Am J Pathol ; 166(1): 275-86, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15632019

ABSTRACT

Human polycystic lipomembraneous osteodysplasia with sclerosing leukoencephalopathy, also known as Nasu-Hakola disease, has been described to be associated with mutations affecting the immunoreceptor tyrosine-based activation motif-bearing KARAP/DAP12 immunoreceptor gene. Patients present bone fragilities and severe neurological alterations leading to presenile dementia. Here we investigated whether the absence of KARAP/DAP12-mediated signals in loss-of-function (KDelta75) mice also leads to bone and central nervous system pathological features. Histological analysis of adult KDelta75 mice brains revealed a diffuse hypomyelination predominating in anterior brain regions. As this was not accompanied by oligodendrocyte degeneration or microglial cell activation it suggests a developmental defect of myelin formation. Interestingly, in postnatal KDelta75 mice, we observed a dramatic reduction in microglial cell numbers similar to in vitro microglial cell differentiation impairment. Our results raise the intriguing possibility that defective microglial cell differentiation might be responsible for abnormal myelin development. Histomorphometry revealed that bone remodeling is also altered, because of a resorption defect, associated with a severe block of in vitro osteoclast differentiation. In addition, we show that, among monocytic lineages, KARAP/DAP12 specifically controls microglial and osteoclast differentiation. Our results confirm that KARAP/DAP12-mediated signals play an important role in the regulation of both brain and bone homeostasis. Yet, important differences exist between the symptoms observed in Nasu-Hakola patients and KDelta75 mice.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Bone Diseases/genetics , Brain Damage, Chronic/genetics , Microglia/pathology , Osteoclasts/pathology , Receptors, Immunologic/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Bone Diseases/pathology , Brain Damage, Chronic/pathology , Mice , Mice, Knockout , Receptors, Immunologic/genetics
20.
Immunity ; 21(3): 367-77, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15357948

ABSTRACT

MICA molecules interact with the NKG2D-activating receptor on human NK and CD8 T cells. We investigated the participation of the MICA/NKG2D pathway in the destruction of intestinal epithelium by intraepithelial T lymphocytes (IEL) in Celiac disease and its premalignant complication, refractory sprue. We show that MICA is strongly expressed at epithelial cell surface in patients with active disease and is induced by gliadin or its p31-49 derived peptide upon in vitro challenge, an effect relayed by IL-15. This triggers direct activation and costimulation of IEL through engagement of NKG2D, leading to an innate-like cytotoxicity toward epithelial targets and enhanced TCR-dependent CD8 T cell-mediated adaptive response. Villous atrophy in Celiac disease might thus be ascribed to an IEL-mediated damage to enterocytes involving NKG2D/MICA interaction after gliadin-induced expression of MICA on gut epithelium. This supports a key role for MIC/NKG2D in the activation of intraepithelial immunity in response to danger.


Subject(s)
Celiac Disease/immunology , Intestinal Mucosa/immunology , Proteins/immunology , Receptors, Immunologic/immunology , Atrophy/immunology , Atrophy/pathology , Celiac Disease/blood , Celiac Disease/pathology , Cytotoxicity Tests, Immunologic , Enterocytes/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gliadin/pharmacology , HeLa Cells , Humans , Immunohistochemistry , Interleukin-15/immunology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestine, Small/immunology , Intestine, Small/pathology , NK Cell Lectin-Like Receptor Subfamily K , Protein Biosynthesis , Proteins/drug effects , RNA, Long Noncoding , RNA, Untranslated , Receptors, Natural Killer Cell , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...