Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Sci Immunol ; 9(93): eadj4775, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38489352

ABSTRACT

The gut microbiota promotes immune system development in early life, but the interactions between the gut metabolome and immune cells in the neonatal gut remain largely undefined. Here, we demonstrate that the neonatal gut is uniquely enriched with neurotransmitters, including serotonin, and that specific gut bacteria directly produce serotonin while down-regulating monoamine oxidase A to limit serotonin breakdown. We found that serotonin directly signals to T cells to increase intracellular indole-3-acetaldehdye and inhibit mTOR activation, thereby promoting the differentiation of regulatory T cells, both ex vivo and in vivo in the neonatal intestine. Oral gavage of serotonin into neonatal mice resulted in long-term T cell-mediated antigen-specific immune tolerance toward both dietary antigens and commensal bacteria. Together, our study has uncovered an important role for specific gut bacteria to increase serotonin availability in the neonatal gut and identified a function of gut serotonin in shaping T cell response to dietary antigens and commensal bacteria to promote immune tolerance in early life.


Subject(s)
Gastrointestinal Microbiome , Serotonin , Animals , Mice , Bacteria , Immune Tolerance , Antigens
2.
Immunity ; 56(12): 2674-2676, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38091947

ABSTRACT

Multiple sclerosis shows a strong sex bias, with unclear mechanisms. In this issue of Immunity, Peng et al. elucidate a female-biased increase in intestinal dopamine signaling that diminishes protective Lactobacillus and exacerbates inflammation in a mouse model of multiple sclerosis.


Subject(s)
Multiple Sclerosis , Animals , Mice , Female , Dopamine , Sexism , Inflammation
3.
bioRxiv ; 2023 Aug 15.
Article in English | MEDLINE | ID: mdl-37645894

ABSTRACT

Despite the success of fructose as a low-cost food additive, recent epidemiological evidence suggests that high fructose consumption by pregnant mothers or during adolescence is associated with disrupted neurodevelopment 1-7 . An essential step in appropriate mammalian neurodevelopment is the synaptic pruning and elimination of newly-formed neurons by microglia, the central nervous system's (CNS) resident professional phagocyte 8-10 . Whether early life high fructose consumption affects microglia function and if this directly impacts neurodevelopment remains unknown. Here, we show that both offspring born to dams fed a high fructose diet and neonates exposed to high fructose exhibit decreased microglial density, increased uncleared apoptotic cells, and decreased synaptic pruning in vivo . Importantly, deletion of the high affinity fructose transporter SLC2A5 (GLUT5) in neonates completely reversed microglia dysfunction, suggesting that high fructose directly affects neonatal development. Mechanistically, we found that high fructose treatment of both mouse and human microglia suppresses synaptic pruning and phagocytosis capacity which is fully reversed in GLUT5-deficient microglia. Using a combination of in vivo and in vitro nuclear magnetic resonance- and mass spectrometry-based fructose tracing, we found that high fructose drives significant GLUT5-dependent fructose uptake and catabolism, rewiring microglia metabolism towards a hypo-phagocytic state. Importantly, mice exposed to high fructose as neonates exhibited cognitive defects and developed anxiety-like behavior which were rescued in GLUT5-deficient animals. Our findings provide a mechanistic explanation for the epidemiological observation that early life high fructose exposure is associated with increased prevalence of adolescent anxiety disorders.

4.
Microorganisms ; 11(3)2023 Feb 27.
Article in English | MEDLINE | ID: mdl-36985171

ABSTRACT

There has been growing interest in the complex host-microbe interactions within the human gut and the role these interactions play in systemic health and disease. As an essential metabolic organ, the liver is intimately coupled to the intestinal microbial environment via the portal venous system. Our understanding of the gut-liver axis comes almost exclusively from studies of adults; the gut-liver axis in children, who have unique physiology and differing gut microbial communities, remains poorly understood. Here, we provide a comprehensive overview of common pediatric hepatobiliary conditions and recent studies exploring the contributions of the gut microbiota to these conditions or changes of the gut microbiota due to these conditions. We examine the current literature regarding the microbial alterations that take place in biliary atresia, pediatric non-alcoholic fatty liver disease, Wilson's disease, cystic fibrosis, inflammatory bowel disease, and viral hepatitis. Finally, we propose potential therapeutic approaches involving modulation of the gut microbiota and the gut-liver axis to mitigate the progression of pediatric liver disease.

5.
Pediatr Res ; 93(5): 1375-1382, 2023 04.
Article in English | MEDLINE | ID: mdl-35986143

ABSTRACT

BACKGROUND: In utero transmission of SARS coronavirus 2 (SARS-CoV-2) has not been fully investigated. We investigated whether newborns of mothers with COVID-19 during pregnancy might harbor SARS-CoV-2 in the gastrointestinal tract. METHODS: This cohort study investigated stool from 14 newborns born at 25-41 weeks admitted at delivery to our urban academic hospital whose mothers had COVID-19 during pregnancy. Eleven mothers had COVID-19 resolved more than 10 weeks before delivery. Newborn stool was evaluated for SARS-CoV-2 RNA, Spike protein, and induction of inflammatory cytokines interleukin-6 (IL-6) and interferon-γ (IFN-γ) in macrophages. RESULTS: Despite negative SARS CoV-2 nasal PCRs from all newborns, viral RNAs and Spike protein were detected in the stool of 11 out of 14 newborns as early as the first day of life and increased over time in 6. Stool homogenates from all 14 newborns elicited elevated inflammatory IL-6 and IFN-γ from macrophages. Most newborns were clinically well except for one death from gestational autoimmune liver disease and another who developed necrotizing enterocolitis. CONCLUSIONS: These findings suggest in utero transmission of SARS-CoV-2 and possible persistent intestinal viral reservoirs in the newborns. Further investigation is required to understand the mechanisms and their clinical implications. IMPACT: SARS-CoV-2 RNAs or Spike protein was detected in the stool of 11 out of 14 preterm newborns born to mothers with resolved COVID-19 weeks prior to delivery despite negative newborn nasal PCR swabs. These novel findings suggest risk of in utero SARS-CoV-2 transmission to the fetal intestine during gestation. The presence of SARS-CoV-2 RNAs and Spike protein in the intestines of newborns may potentially impact the development of the gut microbiome and the immune system; the long-term health impact on the preterm infants should be further investigated.


Subject(s)
COVID-19 , Pregnancy Complications, Infectious , Pregnancy , Female , Infant, Newborn , Humans , SARS-CoV-2 , Cohort Studies , RNA, Viral , Spike Glycoprotein, Coronavirus , Interleukin-6 , Infant, Premature , Pregnancy Complications, Infectious/diagnosis , Infectious Disease Transmission, Vertical
6.
Gut Microbes ; 14(1): 2105609, 2022.
Article in English | MEDLINE | ID: mdl-35915556

ABSTRACT

The gut microbiome is intricately coupled with immune regulation and metabolism, but its role in Coronavirus Disease 2019 (COVID-19) is not fully understood. Severe and fatal COVID-19 is characterized by poor anti-viral immunity and hypercoagulation, particularly in males. Here, we define multiple pathways by which the gut microbiome protects mammalian hosts from SARS-CoV-2 intranasal infection, both locally and systemically, via production of short-chain fatty acids (SCFAs). SCFAs reduced viral burdens in the airways and intestines by downregulating the SARS-CoV-2 entry receptor, angiotensin-converting enzyme 2 (ACE2), and enhancing adaptive immunity via GPR41 and 43 in male animals. We further identify a novel role for the gut microbiome in regulating systemic coagulation response by limiting megakaryocyte proliferation and platelet turnover via the Sh2b3-Mpl axis. Taken together, our findings have unraveled novel functions of SCFAs and fiber-fermenting gut bacteria to dampen viral entry and hypercoagulation and promote adaptive antiviral immunity.


Subject(s)
COVID-19 , Gastrointestinal Microbiome , Animals , Antiviral Agents/therapeutic use , Fatty Acids, Volatile , Male , Mammals/metabolism , Peptidyl-Dipeptidase A/metabolism , SARS-CoV-2
7.
Sci Immunol ; 7(72): eabh3816, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35687695

ABSTRACT

The gut microbiome elicits antigen-specific immunoglobulin G (IgG) at steady state that cross-reacts to pathogens to confer protection against systemic infection. The role of gut microbiome-specific IgG antibodies in the development of the gut microbiome and immunity against enteric pathogens in early life, however, remains largely undefined. In this study, we show that gut microbiome-induced maternal IgG is transferred to the neonatal intestine through maternal milk via the neonatal Fc receptor and directly inhibits Citrobacter rodentium colonization and attachment to the mucosa. Enhanced neonatal immunity against oral C. rodentium infection was observed after maternal immunization with a gut microbiome-derived IgG antigen, outer membrane protein A, or induction of IgG-inducing gut bacteria. Furthermore, by generating a gene-targeted mouse model with complete IgG deficiency, we demonstrate that IgG knockout neonates are more susceptible to C. rodentium infection and exhibit alterations of the gut microbiome that promote differentiation of interleukin-17A-producing γδ T cells in the intestine, which persist into adulthood and contribute to increased disease severity in a dextran sulfate sodium-induced mouse model of colitis. Together, our studies have defined a critical role for maternal gut microbiome-specific IgG antibodies in promoting immunity against enteric pathogens and shaping the development of the gut microbiome and immune cells in early life.


Subject(s)
Colitis , Enterobacteriaceae Infections , Gastrointestinal Microbiome , Animals , Citrobacter rodentium , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/prevention & control , Immunoglobulin G , Mice
8.
Science ; 373(6558): 967-968, 2021 08 27.
Article in English | MEDLINE | ID: mdl-34446596
9.
Microorganisms ; 8(12)2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33333813

ABSTRACT

Pregnancy induces unique changes in maternal immune responses and metabolism. Drastic physiologic adaptations, in an intricately coordinated fashion, allow the maternal body to support the healthy growth of the fetus. The gut microbiome plays a central role in the regulation of the immune system, metabolism, and resistance to infections. Studies have reported changes in the maternal microbiome in the gut, vagina, and oral cavity during pregnancy; it remains unclear whether/how these changes might be related to maternal immune responses, metabolism, and susceptibility to infections during pregnancy. Our understanding of the concerted adaption of these different aspects of the human physiology to promote a successful pregnant remains limited. Here, we provide a comprehensive documentation and discussion of changes in the maternal microbiome in the gut, oral cavity, and vagina during pregnancy, metabolic changes and complications in the mother and newborn that may be, in part, driven by maternal gut dysbiosis, and, lastly, common infections in pregnancy. This review aims to shed light on how dysregulation of the maternal microbiome may underlie obstetrical metabolic complications and infections.

10.
Curr Opin Microbiol ; 56: 30-37, 2020 08.
Article in English | MEDLINE | ID: mdl-32634598

ABSTRACT

Early life is a critical time window for the neonatal gut to be progressively populated with different bacterial species that collectively promote gut maturation. A fully developed and healthy gut microbiome in neonates is an important driver for the development of other aspects of health. Unlike the relatively stable gut microbiome in adults, the developing gut microbiome in neonates exhibits higher plasticity and adaptability. This also underscores the unique window of opportunity for intervention or preventive measures to improve long-term health through modulations of the gut microbiome in early life. Better understanding of the neonatal gut microbiome - how it arises and how it impacts immune cell development - will help us appreciate the underpinnings of immune-related diseases. Here, we examine recent findings on the neonatal gut microbiome and discuss their implications for understanding this important driver of the maturation of the immune system and immunity against infections in early life.


Subject(s)
Gastrointestinal Microbiome , Infant, Newborn/immunology , Intestines/immunology , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Humans , Immunity , Infant, Newborn/growth & development , Intestines/growth & development , Intestines/microbiology
11.
Cell Host Microbe ; 27(1): 11-13, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31951821

ABSTRACT

The neonatal gut microbiome undergoes dynamic changes in response to many nutritional and environmental variables. A recent study by Singer et al. in Nature Medicine elucidates several mechanisms to inhibit the expansion of gut-derived pathobionts in a dysbiotic neonatal gut and prevent these pathobionts from disseminating systemically and causing sepsis in neonatal mice.


Subject(s)
Gastrointestinal Microbiome , Sepsis , Animals , Animals, Newborn , Dysbiosis , Mice
12.
Nat Commun ; 10(1): 660, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30737385

ABSTRACT

Microfold cells (M-cells) are specialized cells of the intestine that sample luminal microbiota and dietary antigens to educate the immune cells of the intestinal lymphoid follicles. The function of M-cells in systemic inflammatory responses are still unclear. Here we show that epithelial non-canonical NFkB signaling mediated by NFkB-inducing kinase (NIK) is highly active in intestinal lymphoid follicles, and is required for M-cell maintenance. Intestinal NIK signaling modulates M-cell differentiation and elicits both local and systemic IL-17A and IgA production. Importantly, intestinal NIK signaling is active in mouse models of colitis and patients with inflammatory bowel diseases; meanwhile, constitutive NIK signaling increases the susceptibility to inflammatory injury by inducing ectopic M-cell differentiation and a chronic increase of IL-17A. Our work thus defines an important function of non-canonical NFkB and M-cells in immune homeostasis, inflammation and polymicrobial sepsis.


Subject(s)
NF-kappa B/metabolism , Animals , B-Lymphocytes/metabolism , Blotting, Western , Colitis/immunology , Colitis/metabolism , Colon/metabolism , Colon/pathology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunoglobulin A/metabolism , Interleukin-17/metabolism , Intestines/immunology , Mice , Protein Serine-Threonine Kinases , RNA, Ribosomal, 16S/genetics , Sepsis/genetics , Sepsis/metabolism , Signal Transduction/physiology
13.
Cell Host Microbe ; 25(2): 313-323.e4, 2019 02 13.
Article in English | MEDLINE | ID: mdl-30686564

ABSTRACT

Owing to immature immune systems and impaired colonization resistance mediated by the microbiota, infants are more susceptible to enteric infections. Maternal antibodies can provide immunity, with maternal vaccination offering a protective strategy. We find that oral infection of adult females with the enteric pathogen Citrobacter rodentium protects dams and offspring against oral challenge. Parenteral immunization of dams with heat-inactivated C. rodentium reduces pathogen loads and mortality in offspring but not mothers. IgG, but not IgA or IgM, transferred through breast milk to the intestinal lumen of suckling offspring, coats the pathogen and reduces intestinal colonization. Protective IgG largely recognizes virulence factors encoded within the locus of enterocyte effacement (LEE) pathogenicity island, including the adhesin Intimin and T3SS filament EspA, which are major antigens conferring protection. Thus, pathogen-specific IgG in breast milk induced during maternal infection or immunization protects neonates against infection with an attaching and effacing pathogen.


Subject(s)
Antibodies, Bacterial/immunology , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/prevention & control , Immunization, Passive , Immunoglobulin G/immunology , Milk/immunology , Animals , Animals, Newborn , Antigens, Bacterial/immunology , Disease Models, Animal , Immunologic Factors/immunology , Mice , Survival Analysis
14.
Mol Oral Microbiol ; 34(2): 27-38, 2019 04.
Article in English | MEDLINE | ID: mdl-30632295

ABSTRACT

Neutrophils are phagocytic innate immune cells essential for killing bacteria via activation of a wide variety of effector responses and generation of large amounts of reactive oxygen species (ROS). Majority of the ROS in neutrophils is generated by activation of the superoxide-generating enzyme nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. Independent of their anti-microbial function, NADPH oxidase-derived ROS have emerged as key regulators of host immune responses and neutrophilic inflammation. Data from patients with inherited defects in the NADPH oxidase subunit alleles that ablate its enzyme function as well as mouse models demonstrate profound dysregulation of host inflammatory responses, neutrophil hyper-activation and tissue damage in response to microbial ligands or tissue trauma. A large body of literature now demonstrates how oxidants function as essential signaling molecules that are essential for the regulation of neutrophil responses during priming, degranulation, neutrophil extracellular trap formation, and apoptosis, independent of their role in microbial killing. In this review we summarize how NADPH oxidase-derived oxidants modulate neutrophil function in a cell intrinsic manner and regulate host inflammatory responses. In addition, we summarize studies that have elucidated possible roles of oxidants in neutrophilic responses within the oral mucosa and periodontal disease.


Subject(s)
NADPH Oxidases/immunology , NADPH Oxidases/metabolism , Neutrophils/enzymology , Neutrophils/immunology , Neutrophils/metabolism , Animals , Anti-Infective Agents/metabolism , Anti-Infective Agents/pharmacology , Apoptosis , Bacteria/immunology , Bacteria/pathogenicity , Extracellular Traps , Granulomatous Disease, Chronic/immunology , Humans , Immunity, Innate , Inflammation/immunology , Mice , Mouth Mucosa/immunology , NADPH Oxidase 2 , Oxidative Stress , Periodontal Diseases/immunology , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/pharmacology , Respiratory Burst/immunology
15.
Immunol Rev ; 279(1): 70-89, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28856738

ABSTRACT

The intestinal tract of mammals is colonized by a large number of microorganisms including trillions of bacteria that are referred to collectively as the gut microbiota. These indigenous microorganisms have co-evolved with the host in a symbiotic relationship. In addition to metabolic benefits, symbiotic bacteria provide the host with several functions that promote immune homeostasis, immune responses, and protection against pathogen colonization. The ability of symbiotic bacteria to inhibit pathogen colonization is mediated via several mechanisms including direct killing, competition for limited nutrients, and enhancement of immune responses. Pathogens have evolved strategies to promote their replication in the presence of the gut microbiota. Perturbation of the gut microbiota structure by environmental and genetic factors increases the risk of pathogen infection, promotes the overgrowth of harmful pathobionts, and the development of inflammatory disease. Understanding the interaction of the microbiota with pathogens and the immune system will provide critical insight into the pathogenesis of disease and the development of strategies to prevent and treat inflammatory disease.


Subject(s)
Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Immunity , Inflammation/microbiology , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Animals , Homeostasis , Humans , Intestinal Mucosa/microbiology , Symbiosis
16.
J Immunol ; 199(4): 1362-1371, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28674182

ABSTRACT

Enteric pathogens including Salmonella enteric serovar Typhimurium can breach the epithelial barrier of the host and spread to systemic tissues. In response to infection, the host activates innate immune receptors via the signaling molecule MyD88, which induces protective inflammatory and antimicrobial responses. Most of these innate immune responses have been studied in hematopoietic cells, but the role of MyD88 signaling in other cell types remains poorly understood. Surprisingly, we found that Dermo1-Cre;Myd88fl/fl mice with mesenchymal cell-specific deficiency of MyD88 were less susceptible to orogastric and i.p. STyphimurium infection than their Myd88fl/fl littermates. The reduced susceptibility of Dermo1-Cre;Myd88fl/fl mice to infection was associated with lower loads of S. Typhimurium in the liver and spleen. Mutant analyses revealed that S. Typhimurium employs its virulence type III secretion system 2 to promote its growth through MyD88 signaling pathways in mesenchymal cells. Inflammatory monocytes function as a major cell population for systemic dissemination of S. Typhimurium Mechanistically, mesenchymal cell-specific MyD88 signaling promoted CCL2 production in the liver and spleen and recruitment of inflammatory monocytes to systemic organs in response to STyphimurium infection. Consistently, MyD88 signaling in mesenchymal cells enhanced the number of phagocytes including Ly6ChiLy6G- inflammatory monocytes harboring STyphimurium in the liver. These results suggest that S. Typhimurium promotes its systemic growth and dissemination through MyD88 signaling pathways in mesenchymal cells.


Subject(s)
Monocytes/immunology , Monocytes/microbiology , Myeloid Differentiation Factor 88/metabolism , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/pathogenicity , Animals , Antigens, Ly/analysis , Bacterial Load , Chemokine CCL2/biosynthesis , Immunity, Innate , Liver/immunology , Liver/microbiology , Liver/pathology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Salmonella typhimurium/immunology , Salmonella typhimurium/metabolism , Signal Transduction , Spleen/immunology , Spleen/microbiology , Type III Secretion Systems/metabolism
17.
Exp Mol Med ; 49(5): e339, 2017 05 26.
Article in English | MEDLINE | ID: mdl-28546562

ABSTRACT

Many benefits provided by the gut microbiota to the host rely on its intricate interactions with host cells. Perturbations of the gut microbiota, termed gut dysbiosis, affect the interplay between the gut microbiota and host cells, resulting in dysregulation of inflammation that contributes to the pathogenesis of chronic inflammatory diseases, including inflammatory bowel disease, multiple sclerosis, allergic asthma and rheumatoid arthritis. In this review, we provide an overview of how gut bacteria modulates host metabolic and immune functions, summarize studies that examined the roles of gut dysbiosis in chronic inflammatory diseases, and finally discuss measures to correct gut dysbiosis as potential therapeutics for chronic inflammatory diseases.


Subject(s)
Gastrointestinal Microbiome/immunology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa , Animals , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/microbiology , Asthma/immunology , Asthma/microbiology , Bacteria/immunology , Dysbiosis/microbiology , Humans , Inflammation/immunology , Inflammation/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Multiple Sclerosis/immunology , Multiple Sclerosis/microbiology
18.
Immunity ; 44(3): 647-658, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26944199

ABSTRACT

The gut microbiota is compartmentalized in the intestinal lumen and induces local immune responses, but it remains unknown whether the gut microbiota can induce systemic response and contribute to systemic immunity. We report that selective gut symbiotic gram-negative bacteria were able to disseminate systemically to induce immunoglobulin G (IgG) response, which primarily targeted gram-negative bacterial antigens and conferred protection against systemic infections by E. coli and Salmonella by directly coating bacteria to promote killing by phagocytes. T cells and Toll-like receptor 4 on B cells were important in the generation of microbiota-specific IgG. We identified murein lipoprotein (MLP), a highly conserved gram-negative outer membrane protein, as a major antigen that induced systemic IgG homeostatically in both mice and humans. Administration of anti-MLP IgG conferred crucial protection against systemic Salmonella infection. Thus, our findings reveal an important function for the gut microbiota in combating systemic infection through the induction of protective IgG.


Subject(s)
Gram-Negative Bacteria/immunology , Gram-Negative Bacterial Infections/immunology , Immunoglobulin G/metabolism , Intestines/immunology , Peptidoglycan/immunology , Animals , Bacterial Load/genetics , Homeostasis/genetics , Host-Pathogen Interactions , Immunoglobulin G/genetics , Intestines/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics
19.
Nature ; 530(7590): 354-7, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26814970

ABSTRACT

Inflammasomes are intracellular protein complexes that drive the activation of inflammatory caspases. So far, four inflammasomes involving NLRP1, NLRP3, NLRC4 and AIM2 have been described that recruit the common adaptor protein ASC to activate caspase-1, leading to the secretion of mature IL-1ß and IL-18 proteins. The NLRP3 inflammasome has been implicated in the pathogenesis of several acquired inflammatory diseases as well as cryopyrin-associated periodic fever syndromes (CAPS) caused by inherited NLRP3 mutations. Potassium efflux is a common step that is essential for NLRP3 inflammasome activation induced by many stimuli. Despite extensive investigation, the molecular mechanism leading to NLRP3 activation in response to potassium efflux remains unknown. Here we report the identification of NEK7, a member of the family of mammalian NIMA-related kinases (NEK proteins), as an NLRP3-binding protein that acts downstream of potassium efflux to regulate NLRP3 oligomerization and activation. In the absence of NEK7, caspase-1 activation and IL-1ß release were abrogated in response to signals that activate NLRP3, but not NLRC4 or AIM2 inflammasomes. NLRP3-activating stimuli promoted the NLRP3-NEK7 interaction in a process that was dependent on potassium efflux. NLRP3 associated with the catalytic domain of NEK7, but the catalytic activity of NEK7 was shown to be dispensable for activation of the NLRP3 inflammasome. Activated macrophages formed a high-molecular-mass NLRP3-NEK7 complex, which, along with ASC oligomerization and ASC speck formation, was abrogated in the absence of NEK7. NEK7 was required for macrophages containing the CAPS-associated NLRP3(R258W) activating mutation to activate caspase-1. Mouse chimaeras reconstituted with wild-type, Nek7(-/-) or Nlrp3(-/-) haematopoietic cells showed that NEK7 was required for NLRP3 inflammasome activation in vivo. These studies demonstrate that NEK7 is an essential protein that acts downstream of potassium efflux to mediate NLRP3 inflammasome assembly and activation.


Subject(s)
Carrier Proteins/metabolism , Inflammasomes/chemistry , Inflammasomes/metabolism , Potassium/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Biocatalysis , CARD Signaling Adaptor Proteins , Carrier Proteins/chemistry , Carrier Proteins/genetics , Caspase 1/metabolism , Catalytic Domain , Cells, Cultured , Cryopyrin-Associated Periodic Syndromes/genetics , Enzyme Activation , HEK293 Cells , Humans , Interleukin-1beta/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , NIMA-Related Kinases , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Binding , Protein Multimerization , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics
20.
Cell Host Microbe ; 17(5): 617-27, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25936799

ABSTRACT

Virulence factors expressed by enteric bacteria are pivotal for pathogen colonization and induction of intestinal disease, but the mechanisms by which host immunity regulates pathogen virulence are largely unknown. Here we show that specific antibody responses are required for downregulation of virulence gene expression in Citrobacter rodentium, an enteric pathogen that models human infections with attaching-and-effacing bacteria. In the absence of antibodies against the pathogen, phenotypically virulent C. rodentium, accumulated and infected the epithelium and subsequently invaded the lamina propia, causing host lethality. IgG induced after infection recognized virulence factors and bound virulent bacteria within the intestinal lumen, leading to their engulfment by neutrophils, while phenotypically avirulent pathogens remained in the intestinal lumen and were eventually outcompeted by the microbiota. Thus, the interplay of the innate and adaptive immune system selectively targets virulent C. rodentium in the intestinal lumen to promote pathogen eradication and host survival.


Subject(s)
Citrobacter rodentium/growth & development , Citrobacter rodentium/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Immunity, Humoral , Virulence Factors/immunology , Virulence Factors/metabolism , Animals , Antibodies, Bacterial/immunology , Disease Models, Animal , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , Immunoglobulin G/immunology , Mice, Inbred C57BL , Neutrophils/immunology , Neutrophils/microbiology , Phagocytosis , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...