Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 18.215
Filter
Add more filters

Publication year range
1.
Cell ; 187(3): 750-763.e20, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38242132

ABSTRACT

Breastfeeding offers demonstrable benefits to newborns and infants by providing nourishment and immune protection and by shaping the gut commensal microbiota. Although it has been appreciated for decades that breast milk contains complement components, the physiological relevance of complement in breast milk remains undefined. Here, we demonstrate that weanling mice fostered by complement-deficient dams rapidly succumb when exposed to murine pathogen Citrobacter rodentium (CR), whereas pups fostered on complement-containing milk from wild-type dams can tolerate CR challenge. The complement components in breast milk were shown to directly lyse specific members of gram-positive gut commensal microbiota via a C1-dependent, antibody-independent mechanism, resulting in the deposition of the membrane attack complex and subsequent bacterial lysis. By selectively eliminating members of the commensal gut community, complement components from breast milk shape neonate and infant gut microbial composition to be protective against environmental pathogens such as CR.


Subject(s)
Complement System Proteins , Gastrointestinal Microbiome , Milk , Animals , Female , Humans , Infant , Mice , Bacteria , Breast Feeding , Citrobacter rodentium , Complement System Proteins/analysis , Immunologic Factors , Infant Health , Milk, Human , Milk/chemistry , Enterobacteriaceae Infections/immunology
2.
Cell ; 186(16): 3414-3426.e16, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37541198

ABSTRACT

Lateral transduction (LT) is the process by which temperate phages mobilize large sections of bacterial genomes. Despite its importance, LT has only been observed during prophage induction. Here, we report that superantigen-carrying staphylococcal pathogenicity islands (SaPIs) employ a related but more versatile and complex mechanism of gene transfer to drive chromosomal hypermobility while self-transferring with additional virulence genes from the host. We found that after phage infection or prophage induction, activated SaPIs form concatamers in the bacterial chromosome by switching between parallel genomic tracks in replication bubbles. This dynamic life cycle enables SaPIbov1 to piggyback its LT of staphylococcal pathogenicity island vSaα, which encodes an array of genes involved in host-pathogen interactions, allowing both islands to be mobilized intact and transferred in a single infective particle. Our findings highlight previously unknown roles of pathogenicity islands in bacterial virulence and show that their evolutionary impact extends beyond the genes they carry.


Subject(s)
Genomic Islands , Staphylococcus Phages , Staphylococcus , Genome, Bacterial , Staphylococcus/genetics , Staphylococcus/pathogenicity , Virulence , Transduction, Genetic
3.
Cell ; 186(24): 5375-5393.e25, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37995657

ABSTRACT

Itch is an unpleasant sensation that evokes a desire to scratch. The skin barrier is constantly exposed to microbes and their products. However, the role of microbes in itch generation is unknown. Here, we show that Staphylococcus aureus, a bacterial pathogen associated with itchy skin diseases, directly activates pruriceptor sensory neurons to drive itch. Epicutaneous S. aureus exposure causes robust itch and scratch-induced damage. By testing multiple isogenic bacterial mutants for virulence factors, we identify the S. aureus serine protease V8 as a critical mediator in evoking spontaneous itch and alloknesis. V8 cleaves proteinase-activated receptor 1 (PAR1) on mouse and human sensory neurons. Targeting PAR1 through genetic deficiency, small interfering RNA (siRNA) knockdown, or pharmacological blockade decreases itch and skin damage caused by V8 and S. aureus exposure. Thus, we identify a mechanism of action for a pruritogenic bacterial factor and demonstrate the potential of inhibiting V8-PAR1 signaling to treat itch.


Subject(s)
Peptide Hydrolases , Pruritus , Receptor, PAR-1 , Staphylococcal Infections , Staphylococcus aureus , Animals , Humans , Mice , Peptide Hydrolases/metabolism , Pruritus/microbiology , Receptor, PAR-1/metabolism , Staphylococcus aureus/enzymology , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/physiology , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology
4.
Cell ; 184(14): 3794-3811.e19, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34166614

ABSTRACT

The microbiota plays a fundamental role in regulating host immunity. However, the processes involved in the initiation and regulation of immunity to the microbiota remain largely unknown. Here, we show that the skin microbiota promotes the discrete expression of defined endogenous retroviruses (ERVs). Keratinocyte-intrinsic responses to ERVs depended on cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes protein (STING) signaling and promoted the induction of commensal-specific T cells. Inhibition of ERV reverse transcription significantly impacted these responses, resulting in impaired immunity to the microbiota and its associated tissue repair function. Conversely, a lipid-enriched diet primed the skin for heightened ERV- expression in response to commensal colonization, leading to increased immune responses and tissue inflammation. Together, our results support the idea that the host may have co-opted its endogenous virome as a means to communicate with the exogenous microbiota, resulting in a multi-kingdom dialog that controls both tissue homeostasis and inflammation.


Subject(s)
Endogenous Retroviruses/physiology , Homeostasis , Inflammation/microbiology , Inflammation/pathology , Microbiota , Animals , Bacteria/metabolism , Chromosomes, Bacterial/genetics , Diet, High-Fat , Inflammation/immunology , Inflammation/virology , Interferon Type I/metabolism , Keratinocytes/metabolism , Membrane Proteins/metabolism , Mice, Inbred C57BL , Nucleotidyltransferases/metabolism , Retroelements/genetics , Signal Transduction , Skin/immunology , Skin/microbiology , T-Lymphocytes/immunology , Transcription, Genetic
5.
Cell ; 180(3): 454-470.e18, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32004459

ABSTRACT

Metagenomic inferences of bacterial strain diversity and infectious disease transmission studies largely assume a dominant, within-individual haplotype. We hypothesize that within-individual bacterial population diversity is critical for homeostasis of a healthy microbiome and infection risk. We characterized the evolutionary trajectory and functional distribution of Staphylococcus epidermidis-a keystone skin microbe and opportunistic pathogen. Analyzing 1,482 S. epidermidis genomes from 5 healthy individuals, we found that skin S. epidermidis isolates coalesce into multiple founder lineages rather than a single colonizer. Transmission events, natural selection, and pervasive horizontal gene transfer result in population admixture within skin sites and dissemination of antibiotic resistance genes within-individual. We provide experimental evidence for how admixture can modulate virulence and metabolism. Leveraging data on the contextual microbiome, we assess how interspecies interactions can shape genetic diversity and mobile gene elements. Our study provides insights into how within-individual evolution of human skin microbes shapes their functional diversification.


Subject(s)
Evolution, Molecular , Gene Transfer, Horizontal , Host Microbial Interactions/genetics , Microbiota/genetics , Polymorphism, Single Nucleotide , Skin/microbiology , Staphylococcus epidermidis/genetics , Adult , DNA, Bacterial/genetics , Drug Resistance, Bacterial/genetics , Female , Healthy Volunteers , Humans , Male , Middle Aged , Phylogeny , Staphylococcus epidermidis/isolation & purification , Staphylococcus epidermidis/pathogenicity , Virulence/genetics , Young Adult
6.
Cell ; 182(5): 1311-1327.e14, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32888495

ABSTRACT

Staphylococcus aureus bacteremia (SaB) causes significant disease in humans, carrying mortality rates of ∼25%. The ability to rapidly predict SaB patient responses and guide personalized treatment regimens could reduce mortality. Here, we present a resource of SaB prognostic biomarkers. Integrating proteomic and metabolomic techniques enabled the identification of >10,000 features from >200 serum samples collected upon clinical presentation. We interrogated the complexity of serum using multiple computational strategies, which provided a comprehensive view of the early host response to infection. Our biomarkers exceed the predictive capabilities of those previously reported, particularly when used in combination. Last, we validated the biological contribution of mortality-associated pathways using a murine model of SaB. Our findings represent a starting point for the development of a prognostic test for identifying high-risk patients at a time early enough to trigger intensive monitoring and interventions.


Subject(s)
Bacteremia/blood , Bacteremia/mortality , Staphylococcal Infections/blood , Staphylococcal Infections/mortality , Staphylococcus aureus/pathogenicity , Animals , Bacteremia/metabolism , Biomarkers/blood , Biomarkers/metabolism , Disease Models, Animal , Female , Humans , Male , Metabolomics/methods , Mice , Middle Aged , Prognosis , Proteomics/methods , Risk Factors , Staphylococcal Infections/metabolism
7.
Immunity ; 57(1): 124-140.e7, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38157853

ABSTRACT

Natural killer (NK) cells are present in the circulation and can also be found residing in tissues, and these populations exhibit distinct developmental requirements and are thought to differ in terms of ontogeny. Here, we investigate whether circulating conventional NK (cNK) cells can develop into long-lived tissue-resident NK (trNK) cells following acute infections. We found that viral and bacterial infections of the skin triggered the recruitment of cNK cells and their differentiation into Tcf1hiCD69hi trNK cells that share transcriptional similarity with CD56brightTCF1hi NK cells in human tissues. Skin trNK cells arose from interferon (IFN)-γ-producing effector cells and required restricted expression of the transcriptional regulator Blimp1 to optimize Tcf1-dependent trNK cell formation. Upon secondary infection, trNK cells rapidly gained effector function and mediated an accelerated NK cell response. Thus, cNK cells redistribute and permanently position at sites of previous infection via a mechanism promoting tissue residency that is distinct from Hobit-dependent developmental paths of NK cells and ILC1 seeding tissues during ontogeny.


Subject(s)
Coinfection , Humans , Killer Cells, Natural/metabolism , Cell Differentiation
8.
Cell ; 172(4): 784-796.e18, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29358051

ABSTRACT

Mammalian barrier surfaces are constitutively colonized by numerous microorganisms. We explored how the microbiota was sensed by the immune system and the defining properties of such responses. Here, we show that a skin commensal can induce T cell responses in a manner that is restricted to non-classical MHC class I molecules. These responses are uncoupled from inflammation and highly distinct from pathogen-induced cells. Commensal-specific T cells express a defined gene signature that is characterized by expression of effector genes together with immunoregulatory and tissue-repair signatures. As such, non-classical MHCI-restricted commensal-specific immune responses not only promoted protection to pathogens, but also accelerated skin wound closure. Thus, the microbiota can induce a highly physiological and pleiotropic form of adaptive immunity that couples antimicrobial function with tissue repair. Our work also reveals that non-classical MHC class I molecules, an evolutionarily ancient arm of the immune system, can promote homeostatic immunity to the microbiota.


Subject(s)
Adaptive Immunity , Bacteria/immunology , Histocompatibility Antigens Class I/immunology , Microbiota/immunology , Skin/immunology , T-Lymphocytes/immunology , Animals , Gene Expression Regulation/immunology , Histocompatibility Antigens Class I/genetics , Mice , Mice, Transgenic
9.
Cell ; 172(5): 1038-1049.e10, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29456081

ABSTRACT

ß-lactam antibiotics inhibit bacterial cell wall assembly and, under classical microbiological culture conditions that are generally hypotonic, induce explosive cell death. Here, we show that under more physiological, osmoprotective conditions, for various Gram-positive bacteria, lysis is delayed or abolished, apparently because inhibition of class A penicillin-binding protein leads to a block in autolytic activity. Although these cells still then die by other mechanisms, exogenous lytic enzymes, such as lysozyme, can rescue viability by enabling the escape of cell wall-deficient "L-form" bacteria. This protective L-form conversion was also observed in macrophages and in an animal model, presumably due to the production of host lytic activities, including lysozyme. Our results demonstrate the potential for L-form switching in the host environment and highlight the unexpected effects of innate immune effectors, such as lysozyme, on antibiotic activity. Unlike previously described dormant persisters, L-forms can continue to proliferate in the presence of antibiotic.


Subject(s)
Anti-Bacterial Agents/pharmacology , L Forms/drug effects , Muramidase/metabolism , beta-Lactams/pharmacology , Animals , Bacillus subtilis/drug effects , Bacteriolysis/drug effects , Cell Wall/drug effects , Cell Wall/metabolism , Hydrolases/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Microbial Viability/drug effects , Osmoregulation/drug effects , Penicillin G/pharmacology , Penicillin-Binding Proteins , Peptidoglycan/metabolism , Prophages/drug effects , RAW 264.7 Cells
10.
Cell ; 174(2): 259-270.e11, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29937224

ABSTRACT

Many community- and hospital-acquired bacterial infections are caused by antibiotic-resistant pathogens. Methicillin-resistant Staphylococcus aureus (MRSA) predisposes humans to invasive infections that are difficult to eradicate. We designed a closed-loop gene network programming mammalian cells to autonomously detect and eliminate bacterial infections. The genetic circuit contains human Toll-like receptors as the bacterial sensor and a synthetic promoter driving reversible and adjustable expression of lysostaphin, a bacteriolytic enzyme highly lethal to S. aureus. Immunomimetic designer cells harboring this genetic circuit exhibited fast and robust sense-and-destroy kinetics against live staphylococci. When tested in a foreign-body infection model in mice, microencapsulated cell implants prevented planktonic MRSA infection and reduced MRSA biofilm formation by 91%. Notably, this system achieved a 100% cure rate of acute MRSA infections, whereas conventional vancomycin treatment failed. These results suggest that immunomimetic designer cells could offer a therapeutic approach for early detection, prevention, and cure of pathogenic infections in the post-antibiotic era.


Subject(s)
Biomimetics/methods , Methicillin-Resistant Staphylococcus aureus/physiology , Staphylococcal Infections/prevention & control , Alkaline Phosphatase/blood , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Disk Diffusion Antimicrobial Tests , Female , HEK293 Cells , Humans , Lipopolysaccharide Receptors/genetics , Lysostaphin/metabolism , Lysostaphin/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice , Mice, Inbred C57BL , Plasmids/genetics , Plasmids/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/veterinary , Toll-Like Receptor 1/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 6/genetics , Transcription Factor AP-1/metabolism
11.
Immunity ; 56(7): 1561-1577.e9, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37402364

ABSTRACT

Hypodermis is the predominant site of Staphylococcus aureus infections that cause cellulitis. Given the importance of macrophages in tissue remodeling, we examined the hypodermal macrophages (HDMs) and their impact on host susceptibility to infection. Bulk and single-cell transcriptomics uncovered HDM subsets with CCR2-dichotomy. HDM homeostasis required the fibroblast-derived growth factor CSF1, ablation of which abrogated HDMs from the hypodermal adventitia. Loss of CCR2- HDMs resulted in accumulation of the extracellular matrix component, hyaluronic acid (HA). HDM-mediated HA clearance required sensing by the HA receptor, LYVE-1. Cell-autonomous IGF1 was required for accessibility of AP-1 transcription factor motifs that controlled LYVE-1 expression. Remarkably, loss of HDMs or IGF1 limited Staphylococcus aureus expansion via HA and conferred protection against cellulitis. Our findings reveal a function for macrophages in the regulation of HA with an impact on infection outcomes, which may be harnessed to limit the establishment of infection in the hypodermal niche.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Humans , Staphylococcus aureus/physiology , Cellulitis/metabolism , Macrophages/metabolism , Extracellular Matrix
12.
Immunity ; 56(6): 1239-1254.e7, 2023 06 13.
Article in English | MEDLINE | ID: mdl-37028427

ABSTRACT

Early-life establishment of tolerance to commensal bacteria at barrier surfaces carries enduring implications for immune health but remains poorly understood. Here, we showed that tolerance in skin was controlled by microbial interaction with a specialized subset of antigen-presenting cells. More particularly, CD301b+ type 2 conventional dendritic cells (DCs) in neonatal skin were specifically capable of uptake and presentation of commensal antigens for the generation of regulatory T (Treg) cells. CD301b+ DC2 were enriched for phagocytosis and maturation programs, while also expressing tolerogenic markers. In both human and murine skin, these signatures were reinforced by microbial uptake. In contrast to their adult counterparts or other early-life DC subsets, neonatal CD301b+ DC2 highly expressed the retinoic-acid-producing enzyme, RALDH2, the deletion of which limited commensal-specific Treg cell generation. Thus, synergistic interactions between bacteria and a specialized DC subset critically support early-life tolerance at the cutaneous interface.


Subject(s)
Dendritic Cells , Skin , Animals , Mice , Humans , T-Lymphocytes, Regulatory , Immune Tolerance , Aldehyde Oxidoreductases/metabolism
13.
Cell ; 168(5): 789-800.e10, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28235196

ABSTRACT

The molecular basis of the incomplete penetrance of monogenic disorders is unclear. We describe here eight related individuals with autosomal recessive TIRAP deficiency. Life-threatening staphylococcal disease occurred during childhood in the proband, but not in the other seven homozygotes. Responses to all Toll-like receptor 1/2 (TLR1/2), TLR2/6, and TLR4 agonists were impaired in the fibroblasts and leukocytes of all TIRAP-deficient individuals. However, the whole-blood response to the TLR2/6 agonist staphylococcal lipoteichoic acid (LTA) was abolished only in the index case individual, the only family member lacking LTA-specific antibodies (Abs). This defective response was reversed in the patient, but not in interleukin-1 receptor-associated kinase 4 (IRAK-4)-deficient individuals, by anti-LTA monoclonal antibody (mAb). Anti-LTA mAb also rescued the macrophage response in mice lacking TIRAP, but not TLR2 or MyD88. Thus, acquired anti-LTA Abs rescue TLR2-dependent immunity to staphylococcal LTA in individuals with inherited TIRAP deficiency, accounting for incomplete penetrance. Combined TIRAP and anti-LTA Ab deficiencies underlie staphylococcal disease in this patient.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Lipopolysaccharides/metabolism , Membrane Glycoproteins/deficiency , Receptors, Interleukin-1/deficiency , Staphylococcal Infections/genetics , Staphylococcal Infections/immunology , Teichoic Acids/metabolism , Adaptive Immunity , Child , Female , Fibroblasts/metabolism , Humans , Immunity, Innate , Lipopolysaccharides/immunology , Macrophages/immunology , Male , Membrane Glycoproteins/analysis , Membrane Glycoproteins/genetics , Monocytes/metabolism , Myeloid Differentiation Factor 88/metabolism , Pedigree , Phagocytes/metabolism , Point Mutation , Protein Isoforms/analysis , Protein Isoforms/genetics , Receptors, Interleukin-1/analysis , Receptors, Interleukin-1/genetics , Staphylococcal Infections/drug therapy , Teichoic Acids/immunology , Toll-Like Receptor 2/metabolism , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism
14.
Annu Rev Biochem ; 84: 577-601, 2015.
Article in English | MEDLINE | ID: mdl-26034890

ABSTRACT

Staphylococcus aureus is a major human and veterinary pathogen worldwide. Methicillin-resistant S. aureus (MRSA) poses a significant and enduring problem to the treatment of infection by such strains. Resistance is usually conferred by the acquisition of a nonnative gene encoding a penicillin-binding protein (PBP2a), with significantly lower affinity for ß-lactams. This resistance allows cell-wall biosynthesis, the target of ß-lactams, to continue even in the presence of typically inhibitory concentrations of antibiotic. PBP2a is encoded by the mecA gene, which is carried on a distinct mobile genetic element (SCCmec), the expression of which is controlled through a proteolytic signal transduction pathway comprising a sensor protein (MecR1) and a repressor (MecI). Many of the molecular and biochemical mechanisms underlying methicillin resistance in S. aureus have been elucidated, including regulatory events and the structure of key proteins. Here we review recent advances in this area.


Subject(s)
Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/metabolism , Staphylococcal Infections/microbiology , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Humans , Penicillin-Binding Proteins , Staphylococcal Infections/veterinary , beta-Lactam Resistance
15.
Mol Cell ; 82(5): 907-919.e7, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35134339

ABSTRACT

Prokaryotic organisms have developed multiple defense systems against phages; however, little is known about whether and how these interact with each other. Here, we studied the connection between two of the most prominent prokaryotic immune systems: restriction-modification and CRISPR. While both systems employ enzymes that cleave a specific DNA sequence of the invader, CRISPR nucleases are programmed with phage-derived spacer sequences, which are integrated into the CRISPR locus upon infection. We found that restriction endonucleases provide a short-term defense, which is rapidly overcome through methylation of the phage genome. In a small fraction of the cells, however, restriction results in the acquisition of spacer sequences from the cleavage site, which mediates a robust type II-A CRISPR-Cas immune response against the methylated phage. This mechanism is reminiscent of eukaryotic immunity in which the innate response offers a first temporary line of defense and also activates a second and more robust adaptive response.


Subject(s)
Bacteriophages , DNA, Viral , Bacteriophages/metabolism , CRISPR-Cas Systems , DNA Restriction Enzymes/genetics , DNA, Viral/genetics , Endonucleases/genetics , Immunity
16.
Immunity ; 53(4): 793-804.e9, 2020 10 13.
Article in English | MEDLINE | ID: mdl-32910906

ABSTRACT

Allergies are considered to represent mal-directed type 2 immune responses against mostly innocuous exogenous compounds. Immunoglobulin E (IgE) antibodies are a characteristic feature of allergies and mediate hypersensitivity against allergens through activation of effector cells, particularly mast cells (MCs). Although the physiological functions of this dangerous branch of immunity have remained enigmatic, recent evidence shows that allergic immune reactions can help to protect against the toxicity of venoms. Because bacteria are a potent alternative source of toxins, we assessed the possible role of allergy-like type 2 immunity in antibacterial host defense. We discovered that the adaptive immune response against Staphylococcus aureus (SA) skin infection substantially improved systemic host defense against secondary SA infections in mice. Moreover, this acquired protection depended on IgE effector mechanisms and MCs. Importantly, our results reveal a previously unknown physiological function of allergic immune responses, IgE antibodies, and MCs in host defense against a pathogenic bacterium.


Subject(s)
Adaptive Immunity/immunology , Immunoglobulin E/immunology , Mast Cells/immunology , Staphylococcal Infections/immunology , Staphylococcal Skin Infections/immunology , Staphylococcus aureus/immunology , Allergens/immunology , Animals , Female , Hypersensitivity/immunology , Hypersensitivity/microbiology , Mast Cells/microbiology , Mice , Mice, Inbred C57BL , Skin/immunology , Skin/microbiology , Staphylococcal Infections/microbiology , Staphylococcal Skin Infections/microbiology
17.
Immunity ; 50(1): 121-136.e5, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30594464

ABSTRACT

Dermal fibroblasts (dFBs) resist infection by locally differentiating into adipocytes and producing cathelicidin antimicrobial peptide in response to Staphylococcus aureus (S. aureus). Here, we show that neonatal skin was enriched with adipogenic dFBs and immature dermal fat that highly expressed cathelicidin. The pool of adipogenic and antimicrobial dFBs declined after birth, leading to an age-dependent loss of dermal fat and a decrease in adipogenesis and cathelidicin production in response to infection. Transforming growth factor beta (TGF-ß), which acted on uncommitted embryonic and adult dFBs and inhibited their adipogenic and antimicrobial function, was identified as a key upstream regulator of this process. Furthermore, inhibition of the TGF-ß receptor restored the adipogenic and antimicrobial function of dFBs in culture and increased resistance of adult mice to S. aureus infection. These results provide insight into changes that occur in the skin innate immune system between the perinatal and adult periods of life.


Subject(s)
Aging/immunology , Fibroblasts/physiology , Skin/metabolism , Staphylococcal Infections/immunology , Staphylococcus aureus/physiology , Subcutaneous Fat/metabolism , Transforming Growth Factor beta/metabolism , Adipocytes/metabolism , Adipogenesis , Animals , Anti-Infective Agents/metabolism , Antimicrobial Cationic Peptides/metabolism , Cells, Cultured , Embryo, Mammalian , Humans , Immunity, Innate , Mice , Cathelicidins
18.
EMBO J ; 42(11): e112140, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37038972

ABSTRACT

Unregulated cell cycle progression may have lethal consequences and therefore, bacteria have various mechanisms in place for the precise spatiotemporal control of cell cycle events. We have uncovered a new link between chromosome replication/segregation and splitting of the division septum. We show that the DNA translocase domain-containing divisome protein FtsK regulates cellular levels of a peptidoglycan hydrolase Sle1, which is involved in cell separation in the bacterial pathogen Staphylococcus aureus. FtsK interacts with a chaperone (trigger factor, TF) and establishes a FtsK-dependent TF concentration gradient that is higher in the septal region. Trigger factor binds Sle1 and promotes its preferential export at the septal region, while also preventing Sle1 degradation by the ClpXP proteolytic machinery. Upon conditions that lead to paused septum synthesis, such as DNA damage or impaired DNA replication/segregation, TF gradient is dissipated and Sle1 levels are reduced, thus halting premature septum splitting.


Subject(s)
Escherichia coli Proteins , Staphylococcal Infections , Humans , Chromosome Segregation , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Membrane Proteins/metabolism , Cell Division , Escherichia coli Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Chromosomes, Bacterial/genetics
19.
Immunity ; 48(5): 963-978.e3, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29768179

ABSTRACT

Regulated antimicrobial peptide expression in the intestinal epithelium is key to defense against infection and to microbiota homeostasis. Understanding the mechanisms that regulate such expression is necessary for understanding immune homeostasis and inflammatory disease and for developing safe and effective therapies. We used Caenorhabditis elegans in a preclinical approach to discover mechanisms of antimicrobial gene expression control in the intestinal epithelium. We found an unexpected role for the cholinergic nervous system. Infection-induced acetylcholine release from neurons stimulated muscarinic signaling in the epithelium, driving downstream induction of Wnt expression in the same tissue. Wnt induction activated the epithelial canonical Wnt pathway, resulting in the expression of C-type lectin and lysozyme genes that enhanced host defense. Furthermore, the muscarinic and Wnt pathways are linked by conserved transcription factors. These results reveal a tight connection between the nervous system and the intestinal epithelium, with important implications for host defense, immune homeostasis, and cancer.


Subject(s)
Acetylcholine/immunology , Caenorhabditis elegans/immunology , Intestinal Mucosa/immunology , Wnt Signaling Pathway/immunology , Acetylcholine/metabolism , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/metabolism , Bacteria/immunology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/microbiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/immunology , Caenorhabditis elegans Proteins/metabolism , Gene Expression/immunology , Homeostasis/genetics , Homeostasis/immunology , Host-Pathogen Interactions/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Neurons/immunology , Neurons/metabolism , Wnt Signaling Pathway/genetics
20.
Proc Natl Acad Sci U S A ; 121(22): e2402764121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38771879

ABSTRACT

Staphylococcus aureus (S. aureus) can evade antibiotics and host immune defenses by persisting within infected cells. Here, we demonstrate that in infected host cells, S. aureus type VII secretion system (T7SS) extracellular protein B (EsxB) interacts with the stimulator of interferon genes (STING) protein and suppresses the inflammatory defense mechanism of macrophages during early infection. The binding of EsxB with STING disrupts the K48-linked ubiquitination of EsxB at lysine 33, thereby preventing EsxB degradation. Furthermore, EsxB-STING binding appears to interrupt the interaction of 2 vital regulatory proteins with STING: aspartate-histidine-histidine-cysteine domain-containing protein 3 (DHHC3) and TNF receptor-associated factor 6. This persistent dual suppression of STING interactions deregulates intracellular proinflammatory pathways in macrophages, inhibiting STING's palmitoylation at cysteine 91 and its K63-linked ubiquitination at lysine 83. These findings uncover an immune-evasion mechanism by S. aureus T7SS during intracellular macrophage infection, which has implications for developing effective immunomodulators to combat S. aureus infections.


Subject(s)
Bacterial Proteins , Macrophages , Membrane Proteins , Staphylococcal Infections , Staphylococcus aureus , Type VII Secretion Systems , Ubiquitination , Staphylococcus aureus/immunology , Membrane Proteins/metabolism , Membrane Proteins/immunology , Humans , Bacterial Proteins/metabolism , Bacterial Proteins/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Animals , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcal Infections/metabolism , Type VII Secretion Systems/metabolism , Type VII Secretion Systems/immunology , Type VII Secretion Systems/genetics , Mice , Immune Evasion , Host-Pathogen Interactions/immunology
SELECTION OF CITATIONS
SEARCH DETAIL