Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Immunity ; 52(2): 357-373.e9, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32049051

ABSTRACT

Clearance of apoptotic cells by macrophages prevents excessive inflammation and supports immune tolerance. Here, we examined the effect of blocking apoptotic cell clearance on anti-tumor immune response. We generated an antibody that selectively inhibited efferocytosis by phagocytic receptor MerTK. Blockade of MerTK resulted in accumulation of apoptotic cells within tumors and triggered a type I interferon response. Treatment of tumor-bearing mice with anti-MerTK antibody stimulated T cell activation and synergized with anti-PD-1 or anti-PD-L1 therapy. The anti-tumor effect induced by anti-MerTK treatment was lost in Stinggt/gt mice, but not in Cgas-/- mice. Abolishing cGAMP production in Cgas-/- tumor cells, depletion of extracellular ATP, or inactivation of the ATP-gated P2X7R channel also compromised the effects of MerTK blockade. Mechanistically, extracellular ATP acted via P2X7R to enhance the transport of extracellular cGAMP into macrophages and subsequent STING activation. Thus, MerTK blockade increases tumor immunogenicity and potentiates anti-tumor immunity, which has implications for cancer immunotherapy.


Subject(s)
Macrophages/immunology , Membrane Proteins/metabolism , Neoplasms/immunology , Nucleotides, Cyclic/metabolism , Receptors, Purinergic P2X7/metabolism , c-Mer Tyrosine Kinase/immunology , Adenosine Triphosphate/metabolism , Animals , Apoptosis , B7-H1 Antigen/immunology , Cells, Cultured , Female , Immunity, Innate , Immunotherapy , Interferon Type I/metabolism , Macrophages/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Nucleotidyltransferases/deficiency , Nucleotidyltransferases/metabolism , Phagocytosis , Programmed Cell Death 1 Receptor/immunology , Receptors, Purinergic P2X7/deficiency , Signal Transduction/immunology , Xenograft Model Antitumor Assays , c-Mer Tyrosine Kinase/genetics
2.
Nature ; 574(7777): 249-253, 2019 10.
Article in English | MEDLINE | ID: mdl-31578523

ABSTRACT

The integrity of the mammalian epidermis depends on a balance of proliferation and differentiation in the resident population of stem cells1. The kinase RIPK4 and the transcription factor IRF6 are mutated in severe developmental syndromes in humans, and mice lacking these genes display epidermal hyperproliferation and soft-tissue fusions that result in neonatal lethality2-5. Our understanding of how these genes control epidermal differentiation is incomplete. Here we show that the role of RIPK4 in mouse development requires its kinase activity; that RIPK4 and IRF6 expressed in the epidermis regulate the same biological processes; and that the phosphorylation of IRF6 at Ser413 and Ser424 primes IRF6 for activation. Using RNA sequencing (RNA-seq), histone chromatin immunoprecipitation followed by sequencing (ChIP-seq) and assay for transposase-accessible chromatin using sequencing (ATAC-seq) of skin in wild-type and IRF6-deficient mouse embryos, we define the transcriptional programs that are regulated by IRF6 during epidermal differentiation. IRF6 was enriched at bivalent promoters, and IRF6 deficiency caused defective expression of genes that are involved in the metabolism of lipids and the formation of tight junctions. Accordingly, the lipid composition of the stratum corneum of Irf6-/- skin was abnormal, culminating in a severe defect in the function of the epidermal barrier. Collectively, our results explain how RIPK4 and IRF6 function to ensure the integrity of the epidermis and provide mechanistic insights into why developmental syndromes that are characterized by orofacial, skin and genital abnormalities result when this axis goes awry.


Subject(s)
Cell Differentiation , Epidermal Cells/cytology , Epidermis/physiology , Interferon Regulatory Factors/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Abnormalities, Multiple/genetics , Animals , Cleft Lip/genetics , Cleft Palate/genetics , Cysts/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Epidermal Cells/metabolism , Epidermis/embryology , Eye Abnormalities/genetics , Female , Fingers/abnormalities , Gene Expression Regulation , Interferon Regulatory Factors/deficiency , Interferon Regulatory Factors/genetics , Knee/abnormalities , Knee Joint/abnormalities , Lip/abnormalities , Lipid Metabolism/genetics , Lower Extremity Deformities, Congenital/genetics , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Phosphoserine/metabolism , Protein Serine-Threonine Kinases/genetics , Syndactyly/genetics , Urogenital Abnormalities/genetics
3.
J Immunol ; 208(12): 2632-2642, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35675956

ABSTRACT

Genetic and environmental cues shape the evolution of the B cell Ig repertoire. Activation-induced cytidine deaminase (AID) is essential to generating Ig diversity through isotype class switching and somatic mutations, which then directly influence clonal selection. Impaired B cell development in AID-knockout mice has made it difficult to study Ig diversification in an aging repertoire. Therefore, in this report, we used a novel inducible AID-knockout mouse model and discovered that deleting AID in adult mice caused spontaneous germinal center formation. Deep sequencing of the IgH repertoire revealed that Ab diversification begins early in life and evolves over time. Our data suggest that activated B cells form germinal centers at steady state and facilitate continuous diversification of the B cell repertoire. In support, we identified shared B cell lineages that were class switched and showed age-dependent rates of mutation. Our data provide novel context to the genesis of the B cell repertoire that may benefit the understanding of autoimmunity and the strength of an immune response to infection.


Subject(s)
Cytidine Deaminase , Immunoglobulin Class Switching , Animals , B-Lymphocytes , Cytidine Deaminase/genetics , Germinal Center , Immunoglobulin Class Switching/genetics , Mice , Mice, Knockout , Somatic Hypermutation, Immunoglobulin
4.
Nature ; 559(7712): 120-124, 2018 07.
Article in English | MEDLINE | ID: mdl-29950720

ABSTRACT

OTULIN (OTU deubiquitinase with linear linkage specificity) removes linear polyubiquitin from proteins that have been modified by LUBAC (linear ubiquitin chain assembly complex) and is critical for preventing auto-inflammatory disease1,2 and embryonic lethality during mouse development3. Here we show that OTULIN promotes rather than counteracts LUBAC activity by preventing its auto-ubiquitination with linear polyubiquitin. Thus, knock-in mice that express catalytically inactive OTULIN, either constitutively or selectively in endothelial cells, resembled LUBAC-deficient mice4 and died midgestation as a result of cell death mediated by TNFR1 (tumour necrosis factor receptor 1) and the kinase activity of RIPK1 (receptor-interacting protein kinase 1). Inactivation of OTULIN in adult mice also caused pro-inflammatory cell death. Accordingly, embryonic lethality and adult auto-inflammation were prevented by the combined loss of cell death mediators: caspase 8 for apoptosis and RIPK3 for necroptosis. Unexpectedly, OTULIN mutant mice that lacked caspase 8 and RIPK3 died in the perinatal period, exhibiting enhanced production of type I interferon that was dependent on RIPK1. Collectively, our results indicate that OTULIN and LUBAC function in a linear pathway, and highlight a previously unrecognized interaction between linear ubiquitination, regulators of cell death, and induction of type I interferon.


Subject(s)
Cell Death , Deubiquitinating Enzymes/metabolism , Endopeptidases/metabolism , Inflammation/metabolism , Ubiquitin/chemistry , Ubiquitin/metabolism , Ubiquitination , Animals , Caspase 8/genetics , Caspase 8/metabolism , Cell Death/genetics , Deubiquitinating Enzymes/genetics , Embryo Loss/genetics , Endopeptidases/genetics , Inflammation/enzymology , Inflammation/genetics , Interferon Type I/biosynthesis , Mice , Mice, Inbred C57BL , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Ubiquitination/genetics , Weight Loss/genetics
5.
Nature ; 540(7631): 129-133, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27819682

ABSTRACT

Receptor-interacting protein kinase 1 (RIPK1) promotes cell survival-mice lacking RIPK1 die perinatally, exhibiting aberrant caspase-8-dependent apoptosis and mixed lineage kinase-like (MLKL)-dependent necroptosis. However, mice expressing catalytically inactive RIPK1 are viable, and an ill-defined pro-survival function for the RIPK1 scaffold has therefore been proposed. Here we show that the RIP homotypic interaction motif (RHIM) in RIPK1 prevents the RHIM-containing adaptor protein ZBP1 (Z-DNA binding protein 1; also known as DAI or DLM1) from activating RIPK3 upstream of MLKL. Ripk1RHIM/RHIM mice that expressed mutant RIPK1 with critical RHIM residues IQIG mutated to AAAA died around birth and exhibited RIPK3 autophosphorylation on Thr231 and Ser232, which is a hallmark of necroptosis, in the skin and thymus. Blocking necroptosis with catalytically inactive RIPK3(D161N), RHIM mutant RIPK3, RIPK3 deficiency, or MLKL deficiency prevented lethality in Ripk1RHIM/RHIM mice. Loss of ZBP1, which engages RIPK3 in response to certain viruses but previously had no defined role in development, also prevented perinatal lethality in Ripk1RHIM/RHIM mice. Consistent with the RHIM of RIPK1 functioning as a brake that prevents ZBP1 from engaging the RIPK3 RHIM, ZBP1 interacted with RIPK3 in Ripk1RHIM/RHIMMlkl-/- macrophages, but not in wild-type, Mlkl-/- or Ripk1RHIM/RHIMRipk3RHIM/RHIM macrophages. Collectively, these findings indicate that the RHIM of RIPK1 is critical for preventing ZBP1/RIPK3/MLKL-dependent necroptosis during development.


Subject(s)
Apoptosis , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Glycoproteins/antagonists & inhibitors , Glycoproteins/metabolism , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Vesicular Transport/deficiency , Adaptor Proteins, Vesicular Transport/metabolism , Amino Acid Motifs , Animals , Animals, Newborn , Caspase 8/genetics , Caspase 8/metabolism , Embryo, Mammalian/cytology , Female , Glycoproteins/chemistry , Glycoproteins/deficiency , Macrophages/metabolism , Male , Mice , Mutation , Phosphorylation , Protein Binding , Protein Kinases/deficiency , Protein Kinases/metabolism , RNA-Binding Proteins , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Tumor Necrosis Factor-alpha/pharmacology
6.
Eur J Immunol ; 50(6): 891-902, 2020 06.
Article in English | MEDLINE | ID: mdl-32043568

ABSTRACT

CD96 is a member of the poliovirus receptor (PVR, CD155)-nectin family that includes T cell Ig and ITIM domain (TIGIT) and CD226. While CD96, TIGIT, and CD226 have important roles in regulating NK cell activity, and TIGIT and CD226 have also been shown to regulate T cell responses, it is unclear whether CD96 has inhibitory or stimulatory function in CD8+ T cells. Here, we demonstrate that CD96 has co-stimulatory function on CD8+ T cells. Crosslinking of CD96 on human or mouse CD8+ T cells induced activation, effector cytokine production, and proliferation. CD96 was found to transduce its activating signal through the MEK-ERK pathway. CD96-mediated signaling led to increased frequencies of NUR77- and T-bet-expressing CD8+ T cells and enhanced cytotoxic effector activity, indicating that CD96 can modulate effector T cell differentiation. Antibody blockade of CD96 or genetic ablation of CD96 expression on CD8+ T cells impaired expression of transcription factors and proinflammatory cytokines associated with CD8+ T cell activation in in vivo models. Taken together, CD96 has a co-stimulatory role in CD8+ T cell activation and effector function.


Subject(s)
Antigens, CD/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Lymphocyte Activation , MAP Kinase Signaling System/immunology , Models, Immunological , Animals , Antigens, CD/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Humans , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout
7.
Nat Methods ; 15(7): 512-514, 2018 07.
Article in English | MEDLINE | ID: mdl-29786090

ABSTRACT

Despite widespread use of CRISPR, comprehensive data on the frequency and impact of Cas9-mediated off-targets in modified rodents are limited. Here we present deep-sequencing data from 81 genome-editing projects on mouse and rat genomes at 1,423 predicted off-target sites, 32 of which were confirmed, and show that high-fidelity Cas9 versions reduced off-target mutation rates in vivo. Using whole-genome sequencing data from ten mouse embryos, treated with a single guide RNA (sgRNA), and from their genetic parents, we found 43 off-targets, 30 of which were predicted by an adapted version of GUIDE-seq.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Genetic Engineering , Genomics/methods , Animals , Cell Line , Female , Male , Mice , Multiplex Polymerase Chain Reaction/methods , RNA/genetics , Rats , Whole Genome Sequencing/methods
8.
Nature ; 528(7580): 127-31, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26580007

ABSTRACT

Prevailing dogma holds that cell-cell communication through Notch ligands and receptors determines binary cell fate decisions during progenitor cell divisions, with differentiated lineages remaining fixed. Mucociliary clearance in mammalian respiratory airways depends on secretory cells (club and goblet) and ciliated cells to produce and transport mucus. During development or repair, the closely related Jagged ligands (JAG1 and JAG2) induce Notch signalling to determine the fate of these lineages as they descend from a common proliferating progenitor. In contrast to such situations in which cell fate decisions are made in rapidly dividing populations, cells of the homeostatic adult airway epithelium are long-lived, and little is known about the role of active Notch signalling under such conditions. To disrupt Jagged signalling acutely in adult mammals, here we generate antibody antagonists that selectively target each Jagged paralogue, and determine a crystal structure that explains selectivity. We show that acute Jagged blockade induces a rapid and near-complete loss of club cells, with a concomitant gain in ciliated cells, under homeostatic conditions without increased cell death or division. Fate analyses demonstrate a direct conversion of club cells to ciliated cells without proliferation, meeting a conservative definition of direct transdifferentiation. Jagged inhibition also reversed goblet cell metaplasia in a preclinical asthma model, providing a therapeutic foundation. Our discovery that Jagged antagonism relieves a blockade of cell-to-cell conversion unveils unexpected plasticity, and establishes a model for Notch regulation of transdifferentiation.


Subject(s)
Antibodies/therapeutic use , Cell Transdifferentiation , Lung/cytology , Lung/metabolism , Receptors, Notch/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Asthma/drug therapy , Asthma/metabolism , Asthma/pathology , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/immunology , Calcium-Binding Proteins/metabolism , Cell Death/drug effects , Cell Division/drug effects , Cell Lineage/drug effects , Cell Tracking , Cell Transdifferentiation/drug effects , Cilia/metabolism , Disease Models, Animal , Female , Goblet Cells/cytology , Goblet Cells/drug effects , Goblet Cells/pathology , Homeostasis/drug effects , Humans , Intercellular Signaling Peptides and Proteins/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Jagged-2 Protein , Ligands , Lung/drug effects , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Serrate-Jagged Proteins , Signal Transduction/drug effects
9.
Nature ; 526(7575): 666-71, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26375259

ABSTRACT

Intracellular lipopolysaccharide from Gram-negative bacteria including Escherichia coli, Salmonella typhimurium, Shigella flexneri, and Burkholderia thailandensis activates mouse caspase-11, causing pyroptotic cell death, interleukin-1ß processing, and lethal septic shock. How caspase-11 executes these downstream signalling events is largely unknown. Here we show that gasdermin D is essential for caspase-11-dependent pyroptosis and interleukin-1ß maturation. A forward genetic screen with ethyl-N-nitrosourea-mutagenized mice links Gsdmd to the intracellular lipopolysaccharide response. Macrophages from Gsdmd(-/-) mice generated by gene targeting also exhibit defective pyroptosis and interleukin-1ß secretion induced by cytoplasmic lipopolysaccharide or Gram-negative bacteria. In addition, Gsdmd(-/-) mice are protected from a lethal dose of lipopolysaccharide. Mechanistically, caspase-11 cleaves gasdermin D, and the resulting amino-terminal fragment promotes both pyroptosis and NLRP3-dependent activation of caspase-1 in a cell-intrinsic manner. Our data identify gasdermin D as a critical target of caspase-11 and a key mediator of the host response against Gram-negative bacteria.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Caspases/metabolism , Inflammasomes/metabolism , Signal Transduction , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Caspases, Initiator , Cell Line , Female , Gram-Negative Bacteria/immunology , Humans , Inflammasomes/drug effects , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice , Mutation/genetics , Necrosis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phosphate-Binding Proteins , Protein Processing, Post-Translational/drug effects , Sepsis/microbiology , Signal Transduction/genetics , Survival Analysis
10.
Nature ; 528(7582): 370-5, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26649818

ABSTRACT

Inactivation of the TNFAIP3 gene, encoding the A20 protein, is associated with critical inflammatory diseases including multiple sclerosis, rheumatoid arthritis and Crohn's disease. However, the role of A20 in attenuating inflammatory signalling is unclear owing to paradoxical in vitro and in vivo findings. Here we utilize genetically engineered mice bearing mutations in the A20 ovarian tumour (OTU)-type deubiquitinase domain or in the zinc finger-4 (ZnF4) ubiquitin-binding motif to investigate these discrepancies. We find that phosphorylation of A20 promotes cleavage of Lys63-linked polyubiquitin chains by the OTU domain and enhances ZnF4-mediated substrate ubiquitination. Additionally, levels of linear ubiquitination dictate whether A20-deficient cells die in response to tumour necrosis factor. Mechanistically, linear ubiquitin chains preserve the architecture of the TNFR1 signalling complex by blocking A20-mediated disassembly of Lys63-linked polyubiquitin scaffolds. Collectively, our studies reveal molecular mechanisms whereby A20 deubiquitinase activity and ubiquitin binding, linear ubiquitination, and cellular kinases cooperate to regulate inflammation and cell death.


Subject(s)
Cysteine Endopeptidases/metabolism , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Ubiquitin/chemistry , Ubiquitin/metabolism , Animals , Cell Death , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/genetics , Female , Inflammation/genetics , Inflammation/pathology , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Lysine/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation , Phosphorylation , Polyubiquitin/chemistry , Polyubiquitin/metabolism , Protein Binding , Protein Kinases/metabolism , Signal Transduction , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factor-alpha/metabolism , Ubiquitination
11.
Proc Natl Acad Sci U S A ; 115(50): E11731-E11740, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30504141

ABSTRACT

Natural killer (NK) cell recognition of tumor cells is mediated through activating receptors such as CD226, with suppression of effector functions often controlled by negative regulatory transcription factors such as FOXO1. Here we show that CD226 regulation of NK cell cytotoxicity is facilitated through inactivation of FOXO1. Gene-expression analysis of NK cells isolated from syngeneic tumors grown in wild-type or CD226-deficient mice revealed dysregulated expression of FOXO1-regulated genes in the absence of CD226. In vitro cytotoxicity and stimulation assays demonstrated that CD226 is required for optimal killing of tumor target cells, with engagement of its ligand CD155 resulting in phosphorylation of FOXO1. CD226 deficiency or anti-CD226 antibody blockade impaired cytotoxicity with concomitant compromised inactivation of FOXO1. Furthermore, inhibitors of FOXO1 phosphorylation abrogated CD226-mediated signaling and effector responses. These results define a pathway by which CD226 exerts control of NK cell responses against tumors.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/metabolism , Forkhead Box Protein O1/antagonists & inhibitors , Forkhead Box Protein O1/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Animals , Antigens, Differentiation, T-Lymphocyte/genetics , Cell Line, Tumor , Cytotoxicity, Immunologic , Gene Expression Regulation, Neoplastic , Humans , Ligands , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice , Mice, Knockout , Nectins/metabolism , Phosphorylation , Receptors, Virus/metabolism , Signal Transduction/immunology
12.
Development ; 144(20): 3686-3697, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28893945

ABSTRACT

The Zfp423/ZNF423 gene encodes a 30-zinc-finger transcription factor involved in key developmental pathways. Although null Zfp423 mutants develop cerebellar malformations, the underlying mechanism remains unknown. ZNF423 mutations are associated with Joubert Syndrome, a ciliopathy causing cerebellar vermis hypoplasia and ataxia. ZNF423 participates in the DNA-damage response (DDR), raising questions regarding its role as a regulator of neural progenitor cell cycle progression in cerebellar development. To characterize in vivo the function of ZFP423 in neurogenesis, we analyzed allelic murine mutants in which distinct functional domains are deleted. One deletion impairs mitotic spindle orientation, leading to premature cell cycle exit and Purkinje cell (PC) progenitor pool deletion. The other deletion impairs PC differentiation. In both mutants, cell cycle progression is remarkably delayed and DDR markers are upregulated in cerebellar ventricular zone progenitors. Our in vivo evidence sheds light on the domain-specific roles played by ZFP423 in different aspects of PC progenitor development, and at the same time strengthens the emerging notion that an impaired DDR may be a key factor in the pathogenesis of JS and other ciliopathies.


Subject(s)
Cell Cycle , DNA-Binding Proteins/physiology , Neural Stem Cells/cytology , Neurons/cytology , Purkinje Cells/cytology , Transcription Factors/physiology , Abnormalities, Multiple/genetics , Alleles , Animals , Cell Differentiation , Cell Division , Cell Proliferation , Cerebellum/abnormalities , DNA Damage , Eye Abnormalities/genetics , Gene Deletion , Kidney Diseases, Cystic/genetics , Mice , Mutation , Protein Domains , Retina/abnormalities , Spindle Apparatus/metabolism , Zinc Fingers
13.
Nature ; 509(7500): 366-70, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24739961

ABSTRACT

Lipopolysaccharide from Gram-negative bacteria is sensed in the host cell cytoplasm by a non-canonical inflammasome pathway that ultimately results in caspase-11 activation and cell death. In mouse macrophages, activation of this pathway requires the production of type-I interferons, indicating that interferon-induced genes have a critical role in initiating this pathway. Here we report that a cluster of small interferon-inducible GTPases, the so-called guanylate-binding proteins, is required for the full activity of the non-canonical caspase-11 inflammasome during infections with vacuolar Gram-negative bacteria. We show that guanylate-binding proteins are recruited to intracellular bacterial pathogens and are necessary to induce the lysis of the pathogen-containing vacuole. Lysis of the vacuole releases bacteria into the cytosol, thus allowing the detection of their lipopolysaccharide by a yet unknown lipopolysaccharide sensor. Moreover, recognition of the lysed vacuole by the danger sensor galectin-8 initiates the uptake of bacteria into autophagosomes, which results in a reduction of caspase-11 activation. These results indicate that host-mediated lysis of pathogen-containing vacuoles is an essential immune function and is necessary for efficient recognition of pathogens by inflammasome complexes in the cytosol.


Subject(s)
Caspases/metabolism , GTP Phosphohydrolases/metabolism , Gram-Negative Bacteria/immunology , Inflammasomes/metabolism , Interferon Type I/immunology , Vacuoles/microbiology , Animals , Autophagy/immunology , Caspases, Initiator , Cytosol/microbiology , Enzyme Activation , Galectins/immunology , Gram-Negative Bacteria/growth & development , Gram-Negative Bacteria/pathogenicity , Immunity, Innate/immunology , Inflammasomes/immunology , Lipopolysaccharides/immunology , Mice , Phagosomes/immunology , Phagosomes/microbiology , Salmonella typhimurium/growth & development , Salmonella typhimurium/immunology
14.
Nucleic Acids Res ; 45(8): e62, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28053125

ABSTRACT

Cre/LoxP technology is widely used in the field of mouse genetics for spatial and/or temporal regulation of gene function. For Cre lines generated via pronuclear microinjection of a Cre transgene construct, the integration site is random and in most cases not known. Integration of a transgene can disrupt an endogenous gene, potentially interfering with interpretation of the phenotype. In addition, knowledge of where the transgene is integrated is important for planning of crosses between animals carrying a conditional allele and a given Cre allele in case the alleles are on the same chromosome. We have used targeted locus amplification (TLA) to efficiently map the transgene location in seven previously published Cre and CreERT2 transgenic lines. In all lines, transgene insertion was associated with structural changes of variable complexity, illustrating the importance of testing for rearrangements around the integration site. In all seven lines the exact integration site and breakpoint sequences were identified. Our methods, data and genotyping assays can be used as a resource for the mouse community and our results illustrate the power of the TLA method to not only efficiently map the integration site of any transgene, but also provide additional information regarding the transgene integration events.


Subject(s)
Chromosome Mapping/methods , Genome , Integrases/genetics , Mutagenesis, Insertional , Nucleic Acid Amplification Techniques , Transgenes , Animals , Gene Dosage , Gene Expression , Gene Library , Genetic Loci , High-Throughput Nucleotide Sequencing , Integrases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Spleen/cytology , Spleen/metabolism
15.
Nature ; 478(7368): 255-9, 2011 Sep 18.
Article in English | MEDLINE | ID: mdl-21927002

ABSTRACT

The small intestine epithelium renews every 2 to 5 days, making it one of the most regenerative mammalian tissues. Genetic inducible fate mapping studies have identified two principal epithelial stem cell pools in this tissue. One pool consists of columnar Lgr5-expressing cells that cycle rapidly and are present predominantly at the crypt base. The other pool consists of Bmi1-expressing cells that largely reside above the crypt base. However, the relative functions of these two pools and their interrelationship are not understood. Here we specifically ablated Lgr5-expressing cells in mice using a human diphtheria toxin receptor (DTR) gene knocked into the Lgr5 locus. We found that complete loss of the Lgr5-expressing cells did not perturb homeostasis of the epithelium, indicating that other cell types can compensate for the elimination of this population. After ablation of Lgr5-expressing cells, progeny production by Bmi1-expressing cells increased, indicating that Bmi1-expressing stem cells compensate for the loss of Lgr5-expressing cells. Indeed, lineage tracing showed that Bmi1-expressing cells gave rise to Lgr5-expressing cells, pointing to a hierarchy of stem cells in the intestinal epithelium. Our results demonstrate that Lgr5-expressing cells are dispensable for normal intestinal homeostasis, and that in the absence of these cells, Bmi1-expressing cells can serve as an alternative stem cell pool. These data provide the first experimental evidence for the interrelationship between these populations. The Bmi1-expressing stem cells may represent both a reserve stem cell pool in case of injury to the small intestine epithelium and a source for replenishment of the Lgr5-expressing cells under non-pathological conditions.


Subject(s)
Intestine, Small/cytology , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Repressor Proteins/metabolism , Stem Cells/cytology , Animals , Cell Lineage , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Heparin-binding EGF-like Growth Factor , Homeostasis , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intestine, Small/metabolism , Male , Mice , Mice, Inbred C57BL , Polycomb Repressive Complex 1 , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Regeneration , Stem Cells/metabolism
16.
Nature ; 479(7371): 117-21, 2011 Oct 16.
Article in English | MEDLINE | ID: mdl-22002608

ABSTRACT

Caspase-1 activation by inflammasome scaffolds comprised of intracellular nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs) and the adaptor ASC is believed to be essential for production of the pro-inflammatory cytokines interleukin (IL)-1ß and IL-18 during the innate immune response. Here we show, with C57BL/6 Casp11 gene-targeted mice, that caspase-11 (also known as caspase-4) is critical for caspase-1 activation and IL-1ß production in macrophages infected with Escherichia coli, Citrobacter rodentium or Vibrio cholerae. Strain 129 mice, like Casp11(-/-) mice, exhibited defects in IL-1ß production and harboured a mutation in the Casp11 locus that attenuated caspase-11 expression. This finding is important because published targeting of the Casp1 gene was done using strain 129 embryonic stem cells. Casp1 and Casp11 are too close in the genome to be segregated by recombination; consequently, the published Casp1(-/-) mice lack both caspase-11 and caspase-1. Interestingly, Casp11(-/-) macrophages secreted IL-1ß normally in response to ATP and monosodium urate, indicating that caspase-11 is engaged by a non-canonical inflammasome. Casp1(-/-)Casp11(129mt/129mt) macrophages expressing caspase-11 from a C57BL/6 bacterial artificial chromosome transgene failed to secrete IL-1ß regardless of stimulus, confirming an essential role for caspase-1 in IL-1ß production. Caspase-11 rather than caspase-1, however, was required for non-canonical inflammasome-triggered macrophage cell death, indicating that caspase-11 orchestrates both caspase-1-dependent and -independent outputs. Caspase-1 activation by non-canonical stimuli required NLRP3 and ASC, but caspase-11 processing and cell death did not, implying that there is a distinct activator of caspase-11. Lastly, loss of caspase-11 rather than caspase-1 protected mice from a lethal dose of lipopolysaccharide. These data highlight a unique pro-inflammatory role for caspase-11 in the innate immune response to clinically significant bacterial infections.


Subject(s)
Caspases/metabolism , Inflammasomes/metabolism , Animals , Caspase 1/metabolism , Caspases/genetics , Caspases, Initiator , Citrobacter rodentium/immunology , Enzyme Activation , Escherichia coli/immunology , Immunity, Innate/immunology , Interleukin-1beta/biosynthesis , Interleukin-1beta/metabolism , Lipopolysaccharides/adverse effects , Lipopolysaccharides/immunology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Vibrio cholerae/immunology
18.
Nucleic Acids Res ; 43(19): e124, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26089387

ABSTRACT

A simple and efficient strategy for Bacterial Artificial Chromosome (BAC) recombineering based on co-selection is described. We show that it is possible to efficiently modify two positions of a BAC simultaneously by co-transformation of a single-stranded DNA oligo and a double-stranded selection cassette. The use of co-selection BAC recombineering reduces the DNA manipulation needed to make a conditional knockout gene targeting vector to only two steps: a single round of BAC modification followed by a retrieval step.


Subject(s)
Chromosomes, Artificial, Bacterial , Gene Knockout Techniques/methods , Recombination, Genetic , Genes, Synthetic , Genetic Engineering , Oligonucleotides
19.
Proc Natl Acad Sci U S A ; 111(22): 8209-14, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24843144

ABSTRACT

Cytomegalovirus (CMV) is a widespread opportunistic pathogen that causes birth defects when transmitted transplacentally and severe systemic illness in immunocompromised individuals. MSL-109, a human monoclonal IgG isolated from a CMV seropositive individual, binds to the essential CMV entry glycoprotein H (gH) and prevents infection of cells. Here, we suggest a mechanism for neutralization activity by MSL-109. We define a genetic basis for resistance to MSL-109 and have generated a structural model of gH that reveals the epitope of this neutralizing antibody. Using surface-based, time-resolved FRET, we demonstrate that gH/gL interacts with glycoprotein B (gB). Additionally, we detect homodimers of soluble gH/gL heterodimers and confirm this novel oligomeric assembly on full-length gH/gL expressed on the cell surface. We show that MSL-109 perturbs the dimerization of gH/gL:gH/gL, suggesting that dimerization of gH/gL may be required for infectivity. gH/gL homodimerization may be conserved between alpha- and betaherpesviruses, because both CMV and HSV gH/gL demonstrate self-association in the FRET system. This study provides evidence for a novel mechanism of action for MSL-109 and reveals a previously undescribed aspect of viral entry that may be susceptible to therapeutic intervention.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Antibodies, Viral/immunology , Antibodies, Viral/pharmacology , Base Sequence , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetulus , Dimerization , Drug Resistance, Viral/immunology , Epitope Mapping , Human Umbilical Vein Endothelial Cells , Humans , Molecular Sequence Data , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
20.
J Pathol ; 237(4): 508-19, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26235356

ABSTRACT

CDK8 is a dissociable kinase module of the Mediator complex and has been shown to play an important role in transcriptional regulation in organisms as diverse as yeast and humans. Recent studies suggest that CDK8 functions as an oncoprotein in melanoma and colon cancer. Importantly, these studies were conducted using in vitro cell line models and the role of CDK8 in tumourigenesis in vivo has not been explored. We have generated a mouse with a Cdk8 conditional knockout allele and examined the consequences of Cdk8 loss on normal tissue homeostasis and tumour development in vivo. Cdk8 deletion in the young adult mouse did not induce any gross or histopathological abnormalities, implying that Cdk8 is largely dispensable for somatic cellular homeostasis. In contrast, Cdk8 deletion in the Apc(Min) intestinal tumour model shortened the animals' survival and increased tumour burden. Although Cdk8 deletion did not affect tumour initiation, intestinal tumour size and growth rate were significantly increased in Cdk8-null animals. Transcriptome analysis performed on Cdk8-null intestinal cells revealed up-regulation of genes that are governed by the Polycomb group (PcG) complex. In support of these findings, Cdk8-null intestinal cells and tumours displayed a reduction in histone H3K27 trimethylation, both globally and at the promoters of a number of PcG-regulated genes involved in oncogenic signalling. Together, our findings uncover a tumour suppressor function for CDK8 in vivo and suggest that the role of CDK8 activity in driving oncogenesis is context-specific. Sequencing data were deposited at GEO (Accession No. GSE71385).


Subject(s)
Carcinogenesis/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Cyclin-Dependent Kinase 8/genetics , Gene Expression Regulation, Neoplastic/genetics , Animals , Chromatin Immunoprecipitation , Disease Models, Animal , Enhancer of Zeste Homolog 2 Protein , Fluorescent Antibody Technique , Genes, APC , Genes, Tumor Suppressor , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , Polycomb Repressive Complex 2/metabolism , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL