Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
PLoS Pathog ; 19(9): e1011623, 2023 09.
Article En | MEDLINE | ID: mdl-37703276

The worldwide prevalence of asthma and allergic disorders (allergic rhinitis, atopic dermatitis, food allergy) has been steadily rising in recent decades. It is now estimated that up to 20% of the global population is afflicted by an allergic disease, with increasing incidence rates in both high- and low-income countries. The World Allergy Organization estimates that the total economic burden of asthma and allergic rhinitis alone is approximately $21 billion per year. While allergic stimuli are a complex and heterogenous class of inputs including parasites, pollens, food antigens, drugs, and metals, it has become clear that fungi are major drivers of allergic disease, with estimates that fungal sensitization occurs in 20-30% of atopic individuals and up to 80% of asthma patients. Fungi are eukaryotic microorganisms that can be found throughout the world in high abundance in both indoor and outdoor environments. Understanding how and why fungi act as triggers of allergic type 2 inflammation will be crucial for combating this important health problem. In recent years, there have been significant advances in our understanding of fungi-induced type 2 immunity, however there is still much we don't understand, including why fungi have a tendency to induce allergic reactions in the first place. Here, we will discuss how fungi trigger type 2 immune responses and posit why this response has been evolutionarily selected for induction during fungal encounter.


Asthma , Rhinitis, Allergic , Humans , Inflammation , Eukaryota
2.
Immunity ; 56(7): 1548-1560.e5, 2023 07 11.
Article En | MEDLINE | ID: mdl-37279752

Cryptococcus neoformans is the leading cause of fungal meningitis and is characterized by pathogenic eosinophil accumulation in the context of type-2 inflammation. The chemoattractant receptor GPR35 is expressed by granulocytes and promotes their migration to the inflammatory mediator 5-hydroxyindoleacetic acid (5-HIAA), a serotonin metabolite. Given the inflammatory nature of cryptococcal infection, we examined the role of GPR35 in the circuitry underlying cell recruitment to the lung. GPR35 deficiency dampened eosinophil recruitment and fungal growth, whereas overexpression promoted eosinophil homing to airways and fungal replication. Activated platelets and mast cells were the sources of GPR35 ligand activity and pharmacological inhibition of serotonin conversion to 5-HIAA, or genetic deficiency in 5-HIAA production by platelets and mast cells resulted in more efficient clearance of Cryptococcus. Thus, the 5-HIAA-GPR35 axis is an eosinophil chemoattractant receptor system that modulates the clearance of a lethal fungal pathogen, with implications for the use of serotonin metabolism inhibitors in the treatment of fungal infections.


Cryptococcosis , Invasive Fungal Infections , Humans , Eosinophils , Hydroxyindoleacetic Acid , Mast Cells , Blood Platelets , Ligands , Receptors, Formyl Peptide , Serotonin , Cryptococcosis/microbiology , Cryptococcosis/pathology , Receptors, G-Protein-Coupled/genetics
3.
Cell Host Microbe ; 30(11): 1589-1601.e5, 2022 11 09.
Article En | MEDLINE | ID: mdl-36323314

Candida albicans is the most common cause of fungal infection in humans. IL-17 is critical for defense against superficial fungal infections, but the role of this response in invasive disease is less understood. We show that C. albicans secretes a lipase, Lip2, that facilitates invasive disease via lipid-based suppression of the IL-17 response. Lip2 was identified as an essential virulence factor in a forward genetic screen in a mouse model of bloodstream infection. Murine infection with C. albicans strains lacking Lip2 display exaggerated IL-17 responses that lead to fungal clearance from solid organs and host survival. Both IL-17 signaling and lipase activity are required for Lip2-mediated suppression. Lip2 inhibits IL-17 production indirectly by suppressing IL-23 production by tissue-resident dendritic cells. The lipase hydrolysis product, palmitic acid, similarly suppresses dendritic cell activation in vitro. Thus, C. albicans suppresses antifungal IL-17 defense in solid organs by altering the tissue lipid milieu.


Candida albicans , Interleukin-17 , Humans , Mice , Animals , Candida albicans/genetics , Antifungal Agents/therapeutic use , Lipase/genetics , Lipids , Fungal Proteins
4.
Nature ; 608(7921): 161-167, 2022 08.
Article En | MEDLINE | ID: mdl-35896747

Invasive fungal pathogens are major causes of human mortality and morbidity1,2. Although numerous secreted effector proteins that reprogram innate immunity to promote virulence have been identified in pathogenic bacteria, so far, there are no examples of analogous secreted effector proteins produced by human fungal pathogens. Cryptococcus neoformans, the most common cause of fungal meningitis and a major pathogen in AIDS, induces a pathogenic type 2 response characterized by pulmonary eosinophilia and alternatively activated macrophages3-8. Here, we identify CPL1 as an effector protein secreted by C. neoformans that drives alternative activation (also known as M2 polarization) of macrophages to enable pulmonary infection in mice. We observed that CPL1-enhanced macrophage polarization requires Toll-like receptor 4, which is best known as a receptor for bacterial endotoxin but is also a poorly understood mediator of allergen-induced type 2 responses9-12. We show that this effect is caused by CPL1 itself and not by contaminating lipopolysaccharide. CPL1 is essential for virulence, drives polarization of interstitial macrophages in vivo, and requires type 2 cytokine signalling for its effect on infectivity. Notably, C. neoformans associates selectively with polarized interstitial macrophages during infection, suggesting a mechanism by which C. neoformans generates its own intracellular replication niche within the host. This work identifies a circuit whereby a secreted effector protein produced by a human fungal pathogen reprograms innate immunity, revealing an unexpected role for Toll-like receptor 4 in promoting the pathogenesis of infectious disease.


Cryptococcosis , Cryptococcus neoformans , Fungal Proteins , Hypersensitivity , Inflammation , Toll-Like Receptor 4 , Virulence Factors , Animals , Cryptococcosis/immunology , Cryptococcosis/microbiology , Cryptococcosis/pathology , Cryptococcus neoformans/immunology , Cryptococcus neoformans/pathogenicity , Cytokines/immunology , Fungal Proteins/immunology , Fungal Proteins/metabolism , Hypersensitivity/immunology , Hypersensitivity/microbiology , Immunity, Innate , Inflammation/immunology , Inflammation/microbiology , Lipopolysaccharides/immunology , Lung/immunology , Lung/microbiology , Macrophages/cytology , Macrophages/immunology , Macrophages/microbiology , Mice , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Virulence , Virulence Factors/immunology
5.
Cell Rep ; 35(6): 109105, 2021 05 11.
Article En | MEDLINE | ID: mdl-33979618

Genome engineering of primary human cells with CRISPR-Cas9 has revolutionized experimental and therapeutic approaches to cell biology, but human myeloid-lineage cells have remained largely genetically intractable. We present a method for the delivery of CRISPR-Cas9 ribonucleoprotein (RNP) complexes by nucleofection directly into CD14+ human monocytes purified from peripheral blood, leading to high rates of precise gene knockout. These cells can be efficiently differentiated into monocyte-derived macrophages or dendritic cells. This process yields genetically edited cells that retain transcript and protein markers of myeloid differentiation and phagocytic function. Genetic ablation of the restriction factor SAMHD1 increased HIV-1 infection >50-fold, demonstrating the power of this system for genotype-phenotype interrogation. This fast, flexible, and scalable platform can be used for genetic studies of human myeloid cells in immune signaling, inflammation, cancer immunology, host-pathogen interactions, and beyond, and could facilitate the development of myeloid cellular therapies.


CRISPR-Cas Systems/genetics , Genome/genetics , Myeloid Cells/metabolism , Ribonucleoproteins/metabolism , Animals , Humans , Mice
7.
J Clin Invest ; 130(2): 1036-1051, 2020 02 03.
Article En | MEDLINE | ID: mdl-31961340

Antigen receptor-dependent (AgR-dependent) stimulation of the NF-κB transcription factor in lymphocytes is a required event during adaptive immune response, but dysregulated activation of this signaling pathway can lead to lymphoma. AgR stimulation promotes assembly of the CARMA1-BCL10-MALT1 complex, wherein MALT1 acts as (a) a scaffold to recruit components of the canonical NF-κB machinery and (b) a protease to cleave and inactivate specific substrates, including negative regulators of NF-κB. In multiple lymphoma subtypes, malignant B cells hijack AgR signaling pathways to promote their own growth and survival, and inhibiting MALT1 reduces the viability and growth of these tumors. As such, MALT1 has emerged as a potential pharmaceutical target. Here, we identified G protein-coupled receptor kinase 2 (GRK2) as a new MALT1-interacting protein. We demonstrated that GRK2 binds the death domain of MALT1 and inhibits MALT1 scaffolding and proteolytic activities. We found that lower GRK2 levels in activated B cell-type diffuse large B cell lymphoma (ABC-DLBCL) are associated with reduced survival, and that GRK2 knockdown enhances ABC-DLBCL tumor growth in vitro and in vivo. Together, our findings suggest that GRK2 can function as a tumor suppressor by inhibiting MALT1 and provide a roadmap for developing new strategies to inhibit MALT1-dependent lymphomagenesis.


Carcinogenesis/metabolism , G-Protein-Coupled Receptor Kinase 2/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Oncogene Proteins/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Female , G-Protein-Coupled Receptor Kinase 2/genetics , Humans , Jurkat Cells , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, Inbred NOD , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Oncogene Proteins/genetics
8.
Curr Opin Immunol ; 56: 1-9, 2019 02.
Article En | MEDLINE | ID: mdl-30172069

Proper regulation of sterol biosynthesis is critical for eukaryotic cellular homeostasis. Cholesterol and isoprenoids serve key roles in eukaryotic cells by regulating membrane fluidity and correct localization of proteins. It is becoming increasingly appreciated that dysregulated sterol metabolism engages pathways that lead to inflammation. Of particular importance are inflammasomes, which are multiplatform protein complexes that activate caspase-1 in order to process the pro-inflammatory and pyrogenic cytokines IL-1ß and IL-18. In this review, we highlight recent research that links altered sterol biosynthetic pathway activity to inflammasome activation. We discuss how clues from human genetics have led to new insights into how alterations in isoprenoid biosynthesis connect to inflammation. We also discuss new mechanisms that show how macrophage cholesterol buildup can lead to inflammasome activation.


Inflammasomes/metabolism , Inflammation , Macrophages/physiology , Metabolic Diseases/immunology , Sterols/metabolism , Animals , Caspase 1/metabolism , Homeostasis , Humans , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Metabolic Diseases/genetics , Pyroptosis
9.
Cell ; 171(5): 1057-1071.e11, 2017 Nov 16.
Article En | MEDLINE | ID: mdl-29033131

Type I interferon restrains interleukin-1ß (IL-1ß)-driven inflammation in macrophages by upregulating cholesterol-25-hydroxylase (Ch25h) and repressing SREBP transcription factors. However, the molecular links between lipid metabolism and IL-1ß production remain obscure. Here, we demonstrate that production of 25-hydroxycholesterol (25-HC) by macrophages is required to prevent inflammasome activation by the DNA sensor protein absent in melanoma 2 (AIM2). We find that in response to bacterial infection or lipopolysaccharide (LPS) stimulation, macrophages upregulate Ch25h to maintain repression of SREBP2 activation and cholesterol synthesis. Increasing macrophage cholesterol content is sufficient to trigger IL-1ß release in a crystal-independent but AIM2-dependent manner. Ch25h deficiency results in cholesterol-dependent reduced mitochondrial respiratory capacity and release of mitochondrial DNA into the cytosol. AIM2 deficiency rescues the increased inflammasome activity observed in Ch25h-/-. Therefore, activated macrophages utilize 25-HC in an anti-inflammatory circuit that maintains mitochondrial integrity and prevents spurious AIM2 inflammasome activation.


Cholesterol/metabolism , Inflammasomes/metabolism , Macrophages/metabolism , Animals , Cholesterol/biosynthesis , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/metabolism , Humans , Hydroxycholesterols/metabolism , Inflammasomes/immunology , Inflammation/immunology , Inflammation/pathology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Listeria monocytogenes/physiology , Listeriosis/immunology , Macrophages/cytology , Macrophages/immunology , Membrane Proteins/metabolism , Mice , Mitochondria/metabolism , Oxysterols/metabolism
10.
Sci Immunol ; 2(10)2017 Apr 07.
Article En | MEDLINE | ID: mdl-28738017

Correct positioning of dendritic cells (DCs) is critical for efficient pathogen encounter and antigen presentation. Epstein-Barr virus-induced gene 2 (EBI2) has been identified as a chemoattractant receptor required for naïve CD4+DCIR2+ DC positioning in response to 7α,25-hydroxycholesterol (7α,25-HC). We now provide evidence that a second EBI2 ligand, 7α,27-HC, is involved in splenic DCIR2+ DC positioning and homeostasis. Cyp27a1, the enzyme uniquely required for 7α,27-HC synthesis, is expressed by stromal cells in the region of naïve DC localization. After activation, DCIR2+ DCs move into the T cell zone. We find that EBI2 is rapidly up-regulated in DCIR2+ DCs under certain activation conditions, and positioning at the B-T zone interface depends on EBI2. Under conditions of type I interferon induction, EBI2 ligand levels are elevated, causing activated DCIR2+ DCs to disperse throughout the T zone. Last, we provide evidence that oxysterol metabolism by Batf3-dependent DCs is important for EBI2-dependent positioning of activated DCIR2+ DCs. This work indicates that 7α,27-HC functions as a guidance cue in vivo and reveals a multitiered role for EBI2 in DC positioning. Deficiency in this organizing system results in defective CD4+ T cell responses.

11.
Nat Rev Immunol ; 14(11): 731-43, 2014 Nov.
Article En | MEDLINE | ID: mdl-25324126

Cholesterol and components of the cholesterol biosynthetic pathway have fundamental roles in all mammalian cells. Hydroxylated forms of cholesterol are now emerging as important regulators of immune function. This involves effects on the cholesterol biosynthetic pathway and cell membrane properties, which can have antiviral and anti-inflammatory influences. In addition, a dihydroxylated form of cholesterol functions as an immune cell guidance cue by engaging the G protein-coupled receptor EBI2, and it is required for mounting adaptive immune responses. In this Review, we summarize the current understanding of the closely related oxysterols 25-hydroxycholesterol and 7α,25-dihydroxycholesterol, and the growing evidence that they have wide-ranging influences on innate and adaptive immunity.


Adaptive Immunity , Hydroxycholesterols/immunology , Immunity, Innate , Animals , Humans , Hydroxycholesterols/chemistry , Hydroxycholesterols/metabolism , Mice , Receptors, G-Protein-Coupled/immunology , Sterol Regulatory Element Binding Proteins/metabolism , Viruses/immunology
12.
Science ; 345(6197): 679-84, 2014 Aug 08.
Article En | MEDLINE | ID: mdl-25104388

Type I interferon (IFN) protects against viruses, yet it also has a poorly understood suppressive influence on inflammation. Here, we report that activated mouse macrophages lacking the IFN-stimulated gene cholesterol 25-hydroxylase (Ch25h) and that are unable to produce the oxysterol 25-hydroxycholesterol (25-HC) overproduce inflammatory interleukin-1 (IL-1) family cytokines. 25-HC acts by antagonizing sterol response element-binding protein (SREBP) processing to reduce Il1b transcription and to broadly repress IL-1-activating inflammasomes. In accord with these dual actions of 25-HC, Ch25h-deficient mice exhibit increased sensitivity to septic shock, exacerbated experimental autoimmune encephalomyelitis, and a stronger ability to repress bacterial growth. These findings identify an oxysterol, 25-HC, as a critical mediator in the negative-feedback pathway of IFN signaling on IL-1 family cytokine production and inflammasome activity.


Hydroxycholesterols/metabolism , Inflammation/genetics , Interferon Type I/immunology , Steroid Hydroxylases/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Feedback, Physiological , Inflammasomes/genetics , Inflammasomes/immunology , Inflammation/immunology , Inflammation/microbiology , Interleukin-1/immunology , Macrophage Activation , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Response Elements/genetics , Shock, Septic/genetics , Shock, Septic/immunology , Steroid Hydroxylases/genetics
13.
Cell ; 146(5): 772-84, 2011 Sep 02.
Article En | MEDLINE | ID: mdl-21871655

T cell differentiation into distinct functional effector and inhibitory subsets is regulated, in part, by the cytokine environment present at the time of antigen recognition. Here, we show that hypoxia-inducible factor 1 (HIF-1), a key metabolic sensor, regulates the balance between regulatory T cell (T(reg)) and T(H)17 differentiation. HIF-1 enhances T(H)17 development through direct transcriptional activation of RORγt and via tertiary complex formation with RORγt and p300 recruitment to the IL-17 promoter, thereby regulating T(H)17 signature genes. Concurrently, HIF-1 attenuates T(reg) development by binding Foxp3 and targeting it for proteasomal degradation. Importantly, this regulation occurs under both normoxic and hypoxic conditions. Mice with HIF-1α-deficient T cells are resistant to induction of T(H)17-dependent experimental autoimmune encephalitis associated with diminished T(H)17 and increased T(reg) cells. These findings highlight the importance of metabolic cues in T cell fate determination and suggest that metabolic modulation could ameliorate certain T cell-based immune pathologies.


Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , T-Lymphocytes, Regulatory/cytology , Th17 Cells/cytology , Animals , Base Sequence , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Forkhead Transcription Factors/metabolism , Humans , Hypoxia-Inducible Factor 1/metabolism , Interleukin-17/genetics , Interleukin-17/immunology , Jurkat Cells , Mice , Molecular Sequence Data , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , STAT3 Transcription Factor/metabolism , Sequence Alignment , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , p300-CBP Transcription Factors/metabolism
14.
Science ; 325(5944): 1142-6, 2009 Aug 28.
Article En | MEDLINE | ID: mdl-19696312

CD4+ regulatory T cells (Tregs) maintain immunological self-tolerance and immune homeostasis by suppressing aberrant or excessive immune responses. The core genetic program of Tregs and their ability to suppress pathologic immune responses depends on the transcription factor Foxp3. Despite progress in understanding mechanisms of Foxp3-dependent gene activation, the molecular mechanism of Foxp3-dependent gene repression remains largely unknown. We identified Eos, a zinc-finger transcription factor of the Ikaros family, as a critical mediator of Foxp3-dependent gene silencing in Tregs. Eos interacts directly with Foxp3 and induces chromatin modifications that result in gene silencing in Tregs. Silencing of Eos in Tregs abrogates their ability to suppress immune responses and endows them with partial effector function, thus demonstrating the critical role that Eos plays in Treg programming.


Carrier Proteins/metabolism , Forkhead Transcription Factors/metabolism , Gene Silencing , Nerve Tissue Proteins/metabolism , T-Lymphocytes, Regulatory/physiology , Acetylation , Alcohol Oxidoreductases/metabolism , Animals , Carrier Proteins/genetics , Colitis/immunology , DNA Methylation , DNA-Binding Proteins/metabolism , Gene Knockdown Techniques , Histones/metabolism , Humans , Interleukin-2/biosynthesis , Interleukin-2/genetics , Jurkat Cells , Methylation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , RNA Interference , T-Lymphocytes, Regulatory/immunology , Transduction, Genetic , Zinc Fingers
...