Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Immunity ; 47(2): 339-348.e4, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28801232

ABSTRACT

The gut microbiota regulate susceptibility to multiple human diseases. The Nlrp6-ASC inflammasome is widely regarded as a hallmark host innate immune axis that shapes the gut microbiota composition. This notion stems from studies reporting dysbiosis in mice lacking these inflammasome components when compared with non-littermate wild-type animals. Here, we describe microbial analyses in inflammasome-deficient mice while minimizing non-genetic confounders using littermate-controlled Nlrp6-deficient mice and ex-germ-free littermate-controlled ASC-deficient mice that were all allowed to shape their gut microbiota naturally after birth. Careful microbial phylogenetic analyses of these cohorts failed to reveal regulation of the gut microbiota composition by the Nlrp6- and ASC-dependent inflammasomes. Our results obtained in two geographically separated animal facilities dismiss a generalizable impact of Nlrp6- and ASC-dependent inflammasomes on the composition of the commensal gut microbiota and highlight the necessity for littermate-controlled experimental design in assessing the influence of host immunity on gut microbial ecology.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Bacteria/genetics , Colitis/immunology , Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Inflammasomes/metabolism , Receptors, Cell Surface/metabolism , Animals , CARD Signaling Adaptor Proteins , Cells, Cultured , Colitis/chemically induced , Colitis/microbiology , Dysbiosis/microbiology , Female , Genetic Background , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota , RNA, Ribosomal, 16S/analysis , Receptors, Cell Surface/genetics , Sodium Dodecyl Sulfate
2.
Proc Natl Acad Sci U S A ; 119(2)2022 01 11.
Article in English | MEDLINE | ID: mdl-34996874

ABSTRACT

Lethal toxin (LeTx)-mediated killing of myeloid cells is essential for Bacillus anthracis, the causative agent of anthrax, to establish systemic infection and induce lethal anthrax. The "LeTx-sensitive" NLRP1b inflammasome of BALB/c and 129S macrophages swiftly responds to LeTx intoxication with pyroptosis and secretion of interleukin (IL)-1ß. However, human NLRP1 is nonresponsive to LeTx, prompting us to investigate B. anthracis host-pathogen interactions in C57BL/6J (B6) macrophages and mice that also lack a LeTx-sensitive Nlrp1b allele. Unexpectedly, we found that LeTx intoxication and live B. anthracis infection of B6 macrophages elicited robust secretion of IL-1ß, which critically relied on the NLRP3 inflammasome. TNF signaling through both TNF receptor 1 (TNF-R1) and TNF-R2 were required for B. anthracis-induced NLRP3 inflammasome activation, which was further controlled by RIPK1 kinase activity and LeTx-mediated proteolytic inactivation of MAP kinase signaling. In addition to activating the NLRP3 inflammasome, LeTx-induced MAPKK inactivation and TNF production sensitized B. anthracis-infected macrophages to robust RIPK1- and caspase-8-dependent apoptosis. In agreement, purified LeTx triggered RIPK1 kinase activity- and caspase-8-dependent apoptosis only in macrophages primed with TNF or following engagement of TRIF-dependent Toll-like receptors. Consistently, genetic and pharmacological inhibition of RIPK1 inhibited NLRP3 inflammasome activation and apoptosis of LeTx-intoxicated and B. anthracis-infected macrophages. Caspase-8/RIPK3-deficient mice were significantly protected from B. anthracis-induced lethality, demonstrating the in vivo pathophysiological relevance of this cytotoxic mechanism. Collectively, these results establish TNF- and RIPK1 kinase activity-dependent NLRP3 inflammasome activation and macrophage apoptosis as key host-pathogen mechanisms in lethal anthrax.


Subject(s)
Apoptosis , Bacillus anthracis/metabolism , Caspase 8/metabolism , Inflammasomes/metabolism , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Anthrax , Caspase 8/genetics , Host-Pathogen Interactions/physiology , Inflammasomes/genetics , Interleukin-1beta/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Pyroptosis , Receptor-Interacting Protein Serine-Threonine Kinases , Signal Transduction
3.
EMBO Rep ; 23(10): e54277, 2022 10 06.
Article in English | MEDLINE | ID: mdl-35899491

ABSTRACT

Neutrophils are the most prevalent immune cells in circulation, but the repertoire of canonical inflammasomes in neutrophils and their respective involvement in neutrophil IL-1ß secretion and neutrophil cell death remain unclear. Here, we show that neutrophil-targeted expression of the disease-associated gain-of-function Nlrp3A350V mutant suffices for systemic autoinflammatory disease and tissue pathology in vivo. We confirm the activity of the canonical NLRP3 and NLRC4 inflammasomes in neutrophils, and further show that the NLRP1b, Pyrin and AIM2 inflammasomes also promote maturation and secretion of interleukin (IL)-1ß in cultured bone marrow neutrophils. Notably, all tested canonical inflammasomes promote GSDMD cleavage in neutrophils, and canonical inflammasome-induced pyroptosis and secretion of mature IL-1ß are blunted in GSDMD-knockout neutrophils. In contrast, GSDMD is dispensable for PMA-induced NETosis. We also show that Salmonella Typhimurium-induced pyroptosis is markedly increased in Nox2/Gp91Phox -deficient neutrophils that lack NADPH oxidase activity and are defective in PMA-induced NETosis. In conclusion, we establish the canonical inflammasome repertoire in neutrophils and identify differential roles for GSDMD and the NADPH complex in canonical inflammasome-induced neutrophil pyroptosis and mitogen-induced NETosis, respectively.


Subject(s)
Extracellular Traps , Inflammasomes , Neutrophils , Phosphate-Binding Proteins , Pore Forming Cytotoxic Proteins , Pyroptosis , Animals , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mitogens/metabolism , NADP/metabolism , NADPH Oxidases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Neutrophils/metabolism , Phosphate-Binding Proteins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Pyrin/metabolism
5.
PLoS Biol ; 17(9): e3000354, 2019 09.
Article in English | MEDLINE | ID: mdl-31525186

ABSTRACT

The nucleotide-binding-domain (NBD)-and leucine-rich repeat (LRR)-containing (NLR) family, pyrin-domain-containing 3 (NLRP3) inflammasome drives pathological inflammation in a suite of autoimmune, metabolic, malignant, and neurodegenerative diseases. Additionally, NLRP3 gain-of-function point mutations cause systemic periodic fever syndromes that are collectively known as cryopyrin-associated periodic syndrome (CAPS). There is significant interest in the discovery and development of diarylsulfonylurea Cytokine Release Inhibitory Drugs (CRIDs) such as MCC950/CRID3, a potent and selective inhibitor of the NLRP3 inflammasome pathway, for the treatment of CAPS and other diseases. However, drug discovery efforts have been constrained by the lack of insight into the molecular target and mechanism by which these CRIDs inhibit the NLRP3 inflammasome pathway. Here, we show that the NAIP, CIITA, HET-E, and TP1 (NACHT) domain of NLRP3 is the molecular target of diarylsulfonylurea inhibitors. Interestingly, we find photoaffinity labeling (PAL) of the NACHT domain requires an intact (d)ATP-binding pocket and is substantially reduced for most CAPS-associated NLRP3 mutants. In concordance with this finding, MCC950/CRID3 failed to inhibit NLRP3-driven inflammatory pathology in two mouse models of CAPS. Moreover, it abolished circulating levels of interleukin (IL)-1ß and IL-18 in lipopolysaccharide (LPS)-challenged wild-type mice but not in Nlrp3L351P knock-in mice and ex vivo-stimulated mutant macrophages. These results identify wild-type NLRP3 as the molecular target of MCC950/CRID3 and show that CAPS-related NLRP3 mutants escape efficient MCC950/CRID3 inhibition. Collectively, this work suggests that MCC950/CRID3-based therapies may effectively treat inflammation driven by wild-type NLRP3 but not CAPS-associated mutants.


Subject(s)
Cryopyrin-Associated Periodic Syndromes/genetics , Furans/pharmacology , Inflammasomes/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Cytokines/antagonists & inhibitors , Disease Models, Animal , Drug Evaluation, Preclinical , HEK293 Cells , Heterocyclic Compounds, 4 or More Rings , Humans , Indenes , Lipopolysaccharides , Macrophages/drug effects , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Protein Domains , Sulfones
7.
Immunity ; 35(6): 908-18, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22195746

ABSTRACT

Engagement of tumor necrosis factor receptor 1 signals two diametrically opposed pathways: survival-inflammation and cell death. An additional switch decides, depending on the cellular context, between caspase-dependent apoptosis and RIP kinase (RIPK)-mediated necrosis, also termed necroptosis. We explored the contribution of both cell death pathways in TNF-induced systemic inflammatory response syndrome (SIRS). Deletion of apoptotic executioner caspases (caspase-3 or -7) or inflammatory caspase-1 had no impact on lethal SIRS. However, deletion of RIPK3 conferred complete protection against lethal SIRS and reduced the amounts of circulating damage-associated molecular patterns. Pretreatment with the RIPK1 kinase inhibitor, necrostatin-1, provided a similar effect. These results suggest that RIPK1-RIPK3-mediated cellular damage by necrosis drives mortality during TNF-induced SIRS. RIPK3 deficiency also protected against cecal ligation and puncture, underscoring the clinical relevance of RIPK kinase inhibition in sepsis and identifying components of the necroptotic pathway that are potential therapeutic targets for treatment of SIRS and sepsis.


Subject(s)
Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Systemic Inflammatory Response Syndrome/enzymology , Animals , Apoptosis/drug effects , Caspases/metabolism , Cecal Diseases/genetics , Cecal Diseases/pathology , Gene Deletion , Imidazoles/administration & dosage , Imidazoles/pharmacology , Indoles/administration & dosage , Indoles/pharmacology , Intestinal Mucosa/metabolism , Intestines/drug effects , Intestines/pathology , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Systemic Inflammatory Response Syndrome/genetics , Systemic Inflammatory Response Syndrome/mortality , Tumor Necrosis Factor-alpha/pharmacology
8.
Proc Natl Acad Sci U S A ; 113(50): 14384-14389, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27911804

ABSTRACT

Familial Mediterranean fever (FMF) is the most common monogenic autoinflammatory disease worldwide. It is caused by mutations in the inflammasome adaptor Pyrin, but how FMF mutations alter signaling in FMF patients is unknown. Herein, we establish Clostridium difficile and its enterotoxin A (TcdA) as Pyrin-activating agents and show that wild-type and FMF Pyrin are differentially controlled by microtubules. Diverse microtubule assembly inhibitors prevented Pyrin-mediated caspase-1 activation and secretion of IL-1ß and IL-18 from mouse macrophages and human peripheral blood mononuclear cells (PBMCs). Remarkably, Pyrin inflammasome activation persisted upon microtubule disassembly in PBMCs of FMF patients but not in cells of patients afflicted with other autoinflammatory diseases. We further demonstrate that microtubules control Pyrin activation downstream of Pyrin dephosphorylation and that FMF mutations enable microtubule-independent assembly of apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) micrometer-sized perinuclear structures (specks). The discovery that Pyrin mutations remove the obligatory requirement for microtubules in inflammasome activation provides a conceptual framework for understanding FMF and enables immunological screening of FMF mutations.


Subject(s)
Familial Mediterranean Fever/genetics , Familial Mediterranean Fever/metabolism , Inflammasomes/metabolism , Mutation , Pyrin/genetics , Pyrin/metabolism , Animals , Bacterial Toxins/toxicity , CARD Signaling Adaptor Proteins/metabolism , Clostridium Infections/immunology , Clostridium Infections/metabolism , Enterotoxins/toxicity , Familial Mediterranean Fever/immunology , HEK293 Cells , Humans , Inflammasomes/drug effects , Inflammasomes/immunology , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubules/drug effects , Microtubules/immunology , Microtubules/metabolism , Pyrin/immunology , Tubulin/metabolism
10.
J Biol Chem ; 290(7): 4022-37, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25538244

ABSTRACT

The cytokine TNF is a well known drug target for several inflammatory diseases such as Crohn disease. Despite the great success of TNF blockers, therapy could be improved because of high costs and side effects. Selective inhibition of TNF receptor (TNFR) 1 signaling holds the potential to greatly reduce the pro-inflammatory activity of TNF, thereby preserving the advantageous immunomodulatory signals mediated by TNFR2. We generated a selective human TNFR1 inhibitor based on Nanobody (Nb) technology. Two anti-human TNFR1 Nbs were linked with an anti-albumin Nb to generate Nb Alb-70-96 named "TNF Receptor-One Silencer" (TROS). TROS selectively binds and inhibits TNF/TNFR1 and lymphotoxin-α/TNFR1 signaling with good affinity and IC50 values, both of which are in the nanomolar range. Surface plasmon resonance analysis reveals that TROS competes with TNF for binding to human TNFR1. In HEK293T cells, TROS strongly reduces TNF-induced gene expression, like IL8 and TNF, in a dose-dependent manner; and in ex vivo cultured colon biopsies of CD patients, TROS inhibits inflammation. Finally, in liver chimeric humanized mice, TROS antagonizes inflammation in a model of acute TNF-induced liver inflammation, reflected in reduced human IL8 expression in liver and reduced IL6 levels in serum. These results demonstrate the considerable potential of TROS and justify the evaluation of TROS in relevant disease animal models of both acute and chronic inflammation and eventually in patients.


Subject(s)
Colon/drug effects , Crohn Disease/prevention & control , Inflammation/prevention & control , Liver/drug effects , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Type I/immunology , Single-Domain Antibodies/pharmacology , Amino Acid Sequence , Animals , Colon/immunology , Colon/pathology , Crohn Disease/immunology , Crohn Disease/pathology , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Female , Humans , Inflammation/immunology , Inflammation/pathology , Liver/immunology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Molecular Sequence Data , Protein Conformation , Receptors, Tumor Necrosis Factor, Type I/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Surface Plasmon Resonance , Tumor Necrosis Factor-alpha/pharmacology
11.
Int J Cancer ; 135(3): 742-50, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24382818

ABSTRACT

Tumor necrosis factor (TNF) has remarkable antitumor effects, but its systemic therapeutic use is prevented by its lethal inflammatory effects. TNFR1 (P55) is essential for both the antitumor and toxic effects because both of them are absent in P55-deficient mice. In previous work we demonstrated that P55+/- mice are completely resistant to TNF toxicity, while the antitumor effects induced by TNF combined with interferon gamma (IFNγ) remain fully functional in these mice. Hence, a high dose of TNF/IFNγ has an excellent therapeutic potential when P55 levels are reduced, because TNF induces tumor regression without systemic toxicity. Here, we provide proof of principle for therapeutic application of this approach by using antisense oligonucleotides (ASOs). Treatment of mice with ASOs targeting P55 resulted in a strong reduction in P55 protein levels in liver, small intestine and blood mononuclear cells. This P55 downregulation was associated with significant protection of mice against acute TNF toxicity as measured by hypothermia, systemic inflammation and lethality. This treatment also protected mice against toxicity of TNF/IFNγ treatment in several cancer models: B16Bl6, Lewis lung carcinoma and a lung colony model. Our results confirm the therapeutic value of this strategy, which could lead to the development of a safer and more effective TNF/IFNγ antitumor therapy.


Subject(s)
Carcinoma, Lewis Lung/prevention & control , Disease Models, Animal , Interferon-gamma/toxicity , Melanoma, Experimental/prevention & control , Oligonucleotides, Antisense/pharmacology , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor-alpha/toxicity , Animals , Carcinoma, Lewis Lung/chemically induced , Carcinoma, Lewis Lung/genetics , Female , Intestine, Small/metabolism , Liver/metabolism , Maximum Tolerated Dose , Melanoma, Experimental/chemically induced , Melanoma, Experimental/genetics , Mice , Mice, Inbred C57BL , Mice, Nude , Signal Transduction
12.
J Neurosci ; 32(29): 9805-16, 2012 Jul 18.
Article in English | MEDLINE | ID: mdl-22815495

ABSTRACT

Systemic inflammatory response syndrome (SIRS) is a highly mortal inflammatory disease, associated with systemic inflammation and organ dysfunction. SIRS can have a sterile cause or can be initiated by an infection, called sepsis. The prevalence is high, and available treatments are ineffective and mainly supportive. Consequently, there is an urgent need for new treatments. The brain is one of the first organs affected during SIRS, and sepsis and the consequent neurological complications, such as encephalopathy, are correlated with decreased survival. The choroid plexus (CP) that forms the blood-CSF barrier (BCSFB) is thought to act as a brain "immune sensor" involved in the communication between the peripheral immune system and the CNS. Nevertheless, the involvement of BCSFB integrity in systemic inflammatory diseases is seldom investigated. We report that matrix metalloprotease-8 (MMP8) depletion or inhibition protects mice from death and hypothermia in sepsis and renal ischemia/reperfusion. This effect could be attributed to MMP8-dependent leakage of the BCSFB, caused by collagen cleavage in the extracellular matrix of CP cells, which leads to a dramatic change in cellular morphology. Disruption of the BCSFB results in increased CSF cytokine levels, brain inflammation, and downregulation of the brain glucocorticoid receptor. This receptor is necessary for dampening the inflammatory response. Consequently, MMP8(+/+) mice, in contrast to MMP8(-/-) mice, show no anti-inflammatory response and this results in high mortality. In conclusion, we identify MMP8 as an essential mediator in SIRS and, hence, a potential drug target. We also propose that the mechanism of action of MMP8 involves disruption of the BCSFB integrity.


Subject(s)
Blood-Brain Barrier/metabolism , Endotoxemia/metabolism , Extracellular Matrix/metabolism , Matrix Metalloproteinase 8/metabolism , Systemic Inflammatory Response Syndrome/metabolism , Animals , Brain/metabolism , Choroid Plexus/metabolism , Collagen/metabolism , Corticosterone/blood , Female , Male , Mice , Mice, Knockout
13.
Curr Biol ; 33(1): R33-R36, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36626862

ABSTRACT

A new study reveals how Mycobacterium tuberculosis evades anti-bacterial immunity by modifying the plasma membrane phospholipid composition of infected macrophages, thereby blocking the host's pyroptosis response and supporting chronic infection.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Inflammasomes/metabolism , Tuberculosis/metabolism , Tuberculosis/microbiology , Macrophages/metabolism
14.
J Biol Chem ; 286(30): 26555-67, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21646349

ABSTRACT

As glucocorticoid resistance (GCR) and the concomitant burden pose a worldwide problem, there is an urgent need for a more effective glucocorticoid therapy, for which insights into the molecular mechanisms of GCR are essential. In this study, we addressed the hypothesis that TNFα, a strong pro-inflammatory mediator in numerous inflammatory diseases, compromises the protective function of the glucocorticoid receptor (GR) against TNFα-induced lethal inflammation. Indeed, protection of mice by dexamethasone against TNFα lethality was completely abolished when it was administered after TNFα stimulation, indicating compromised GR function upon TNFα challenge. TNFα-induced GCR was further demonstrated by impaired GR-dependent gene expression in the liver. Furthermore, TNFα down-regulates the levels of both GR mRNA and protein. However, this down-regulation seems to occur independently of GC production, as TNFα also resulted in down-regulation of GR levels in adrenalectomized mice. These findings suggest that the decreased amount of GR determines the GR response and outcome of TNFα-induced shock, as supported by our studies with GR heterozygous mice. We propose that by inducing GCR, TNFα inhibits a major brake on inflammation and thereby amplifies the pro-inflammatory response. Our findings might prove helpful in understanding GCR in inflammatory diseases in which TNFα is intimately involved.


Subject(s)
Down-Regulation , Receptors, Glucocorticoid/biosynthesis , Shock/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Female , Mice , Mice, Transgenic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Glucocorticoid/genetics , Shock/chemically induced , Shock/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/toxicity
15.
Nanoscale ; 13(13): 6592-6604, 2021 Apr 07.
Article in English | MEDLINE | ID: mdl-33885539

ABSTRACT

Inflammasomes are multi-protein complexes that guard against cellular stress and microbial infections. Inflammasome activation studies frequently require delivery of pathogen-derived virulence factors into the cytosol of macrophages and other innate immune cells. This is a challenging requirement since primary macrophages are difficult-to-transfect, especially when it comes to the intracellular delivery of proteins. Here, we report on the use of nanoparticle-sensitized photoporation as a promising upcoming intracellular delivery technology for delivering proteins of various molecular weights into the cytosol of primary macrophages. While 60-70 nm gold nanoparticles are the most commonly used sensitizing nanoparticles for photoporation, here we find that 0.5 µm iron oxide nanoparticles perform markedly better on primary macrophages. We demonstrate that LFn-FlaA or lipopolysaccharides can be delivered in primary macrophages resulting in activation of the NLRC4 or the non-canonical inflammasome, respectively. We furthermore show that photoporation can be used for targeted delivery of these toxins into selected cells, opening up the possibility to study the interaction between inflammasome activated cells and surrounding healthy cells. Taken together, these results show that nanoparticle-sensitized photoporation is very well suited to deliver pathogenic virulence factors in primary macrophages, thus constituting an effective new enabling technology for inflammasome activation studies.


Subject(s)
Inflammasomes , Metal Nanoparticles , Gold , Lipopolysaccharides , Macrophages , Metal Nanoparticles/toxicity
16.
Nat Microbiol ; 4(11): 1805-1814, 2019 11.
Article in English | MEDLINE | ID: mdl-31308522

ABSTRACT

Anthrax is an ancient and deadly disease caused by the spore-forming bacterial pathogen Bacillus anthracis. At present, anthrax mostly affects wildlife and livestock, although it remains a concern for human public health-primarily for people who handle contaminated animal products and as a bioterrorism threat due to the high resilience of spores, a high fatality rate of cases and the lack of a civilian vaccination programme1,2. The cell surface of B. anthracis is covered by a protective paracrystalline monolayer-known as surface layer or S-layer-that is composed of the S-layer proteins Sap or EA1. Here, we generate nanobodies to inhibit the self-assembly of Sap, determine the structure of the Sap S-layer assembly domain (SapAD) and show that the disintegration of the S-layer attenuates the growth of B. anthracis and the pathology of anthrax in vivo. SapAD comprises six ß-sandwich domains that fold and support the formation of S-layers independently of calcium. Sap-inhibitory nanobodies prevented the assembly of Sap and depolymerized existing Sap S-layers in vitro. In vivo, nanobody-mediated disruption of the Sap S-layer resulted in severe morphological defects and attenuated bacterial growth. Subcutaneous delivery of Sap inhibitory nanobodies cleared B. anthracis infection and prevented lethality in a mouse model of anthrax disease. These findings highlight disruption of S-layer integrity as a mechanism that has therapeutic potential in S-layer-carrying pathogens.


Subject(s)
Anthrax/drug therapy , Bacillus anthracis/drug effects , Membrane Glycoproteins/chemistry , Single-Domain Antibodies/administration & dosage , Animals , Anthrax/metabolism , Bacillus anthracis/metabolism , Bacillus anthracis/pathogenicity , Disease Models, Animal , Injections, Subcutaneous , Membrane Glycoproteins/metabolism , Mice , Microbial Viability/drug effects , Models, Molecular , Protein Conformation, beta-Strand/drug effects , Protein Multimerization/drug effects , Single-Domain Antibodies/pharmacology
17.
Arterioscler Thromb Vasc Biol ; 27(7): 1572-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17495235

ABSTRACT

OBJECTIVE: Two types of endothelial progenitor cells (EPCs), early EPCs and late EPCs (also called endothelial outgrowth cells [EOCs]), were described in vitro previously. In this report, we dissect the phenotype of the precursor(s) that generate these cell types with focus on the markers CD34, CD133, and vascular endothelial growth factor receptor-2 (VEGFR2) that have been used to identify putative circulating endothelial precursors. We also included CD45 in the analysis to assess the relation between CD34+ hematopoietic progenitors (HPC), CD34+ endothelial precursors, and both in vitro generated EPC types. Addressing this issue might lead to a better understanding of the lineage and phenotype of the precursor(s) that give rise to both cell types in vitro and may contribute to a consensus on their flowcytometric enumeration. METHODS AND RESULTS: Using cell sorting of human cord blood (UCB) and bone marrow (BM) cells, we demonstrate that EOC generating precursors are confined to a small CD34+ CD45- cell fraction, but not to the CD34+ CD45+ HPC fraction, nor any other CD45+ subpopulation. CD34+ CD45+ HPC generated monocytic cells that displayed characteristics typical for early EPCs. Phenotypic analysis showed that EOC generating CD34+ CD45- cells express VEGFR2 but not CD133, whereas CD34+ CD45+ HPC express CD133 as expected, but not VEGFR2. CONCLUSION: EOCs are not derived from CD133+ cells or CD45+ hematopoietic precursors.


Subject(s)
Antigens, CD/metabolism , Endothelial Cells/cytology , Glycoproteins/metabolism , Hematopoietic Stem Cells/metabolism , Leukocyte Common Antigens/metabolism , Peptides/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , AC133 Antigen , Adolescent , Antigens, CD/genetics , Biomarkers/analysis , Bone Marrow Cells/cytology , Cell Differentiation , Cell Separation , Cells, Cultured , Child , Endothelial Cells/metabolism , Female , Fetal Blood/cytology , Flow Cytometry , Glycoproteins/genetics , Humans , Infant, Newborn , Leukocyte Common Antigens/genetics , Male , Peptides/genetics , Probability , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Vascular Endothelial Growth Factor Receptor-2/genetics
18.
Cell Res ; 32(3): 227-228, 2022 03.
Article in English | MEDLINE | ID: mdl-34934194
19.
Cell Rep ; 21(12): 3427-3444, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29262324

ABSTRACT

The caspase activation and recruitment domain (CARD)-based inflammasome sensors NLRP1b and NLRC4 induce caspase-1-dependent pyroptosis independent of the inflammasome adaptor ASC. Here, we show that NLRP1b and NLRC4 trigger caspase-8-mediated apoptosis as an alternative cell death program in caspase-1-/- macrophages and intestinal epithelial organoids (IECs). The caspase-8 adaptor FADD was recruited to ASC specks, which served as cytosolic platforms for caspase-8 activation and NLRP1b/NLRC4-induced apoptosis. We further found that caspase-1 protease activity dominated over scaffolding functions in suppressing caspase-8 activation and induction of apoptosis of macrophages and IECs. Moreover, TLR-induced c-FLIP expression inhibited caspase-8-mediated apoptosis downstream of ASC speck assembly, but did not affect pyroptosis induction by NLRP1b and NLRC4. Moreover, unlike during pyroptosis, NLRP1b- and NLRC4-elicited apoptosis retained alarmins and the inflammasome-matured cytokines interleukin 1ß (IL-1ß) and IL-18 intracellularly. This work identifies critical mechanisms regulating apoptosis induction by the inflammasome sensors NLRP1b and NLRC4 and suggests converting pyroptosis into apoptosis as a paradigm for suppressing inflammation.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Calcium-Binding Proteins/metabolism , Caspase 1/metabolism , Inflammasomes/metabolism , Pyroptosis , Animals , Caspase 8/metabolism , Enterocytes/metabolism , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL