Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Antimicrob Agents Chemother ; 66(2): e0143121, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34843389

ABSTRACT

Infections caused by Staphylococcus aureus are a leading cause of mortality. Treating infections caused by S. aureus is difficult due to resistance against most traditional antibiotics, including ß-lactams. We previously reported the presence of mutations in gdpP among S. aureus strains that were obtained by serial passaging in ß-lactam drugs. Similar mutations have recently been reported in natural S. aureus isolates that are either nonsusceptible or resistant to ß-lactam antibiotics. gdpP codes for a phosphodiesterase that cleaves cyclic-di-AMP (CDA), a newly discovered second messenger. In this study, we sought to identify the role of gdpP in ß-lactam resistance in S. aureus. Our results showed that gdpP-associated mutations caused loss of phosphodiesterase function, leading to increased CDA accumulation in the bacterial cytosol. Deletion of gdpP led to an enhanced ability of the bacteria to withstand a ß-lactam challenge (2 to 3 log increase in bacterial CFU) by promoting tolerance without enhancing MICs of ß-lactam antibiotics. Our results demonstrated that increased drug tolerance due to loss of GdpP function can provide a selective advantage in acquisition of high-level ß-lactam resistance. Loss of GdpP function thus increases tolerance to ß-lactams that can lead to its therapy failure and can permit ß-lactam resistance to occur more readily.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcus aureus , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Drug Tolerance , Microbial Sensitivity Tests , Staphylococcus aureus/genetics , beta-Lactam Resistance/genetics , beta-Lactams/pharmacology
2.
J Leukoc Biol ; 109(4): 709-715, 2021 04.
Article in English | MEDLINE | ID: mdl-32881103

ABSTRACT

Most modern research into the immune effects of breast milk has focused on the impacts of immunoglobulin or oligosaccharide content. However, immediately prior to parturition, the cell populations of breast milk become selectively enriched for CD8+ T cells of an effector memory subtype. Despite this observation that the cellular content of breast milk contains a distinct leukocyte population when compared to peripheral blood, the physiologic role of these CD8+ effector memory cells is unknown. Research encompassing animal models and humans has demonstrated that leukocytes are capable of transferring antigen-specific immunity even when lysed, dialyzed to enrich for fractions less than 10 kDa, and orally administered. Our previous work built upon these reports to elucidate several aspects of this dialyzable leukocyte extract (DLE) activity: only DLE from T effector memory CD8+ cells was capable of transferring antigen-specific immunity; the DLE activity was TCRß dependent; dendritic cells (DCs) were the cellular target of DLE; and DLE enhanced immune activity in epithelial challenge models via induction of IL-6 from DCs. Herein, we reveal that breast milk dialysate activates similar cytokine and genetic pathways as DLE taken from peripheral blood and murine spleens through TCRß- and CD8-dependent mechanisms. These findings suggest that the CD8+ memory T cells enriched in breast milk, even after potential lysis in the infant gut, may represent a mechanism for passive transfer of cellular immunity from mother to child.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular , Milk, Human/immunology , Animals , Breast Feeding , Cattle , Colostrum/immunology , Cytokines/metabolism , Dialysis , Female , Humans , Immunomodulation , Leukocytes/metabolism , Mice, Inbred C57BL , Models, Animal
3.
Sci Transl Med ; 12(560)2020 09 09.
Article in English | MEDLINE | ID: mdl-32908007

ABSTRACT

Dysbiosis of the skin microbiota is increasingly implicated as a contributor to the pathogenesis of atopic dermatitis (AD). We previously reported first-in-human safety and clinical activity results from topical application of the commensal skin bacterium Roseomonas mucosa for the treatment of AD in 10 adults and 5 children older than 9 years of age. Here, we examined the potential mechanism of action of R. mucosa treatment and its impact on children with AD less than 7 years of age, the most common age group for children with AD. In 15 children with AD, R. mucosa treatment was associated with amelioration of disease severity, improvement in epithelial barrier function, reduced Staphylococcus aureus burden on the skin, and a reduction in topical steroid requirements without severe adverse events. Our observed response rates to R. mucosa treatment were greater than those seen in historical placebo control groups in prior AD studies. Skin improvements and colonization by R. mucosa persisted for up to 8 months after cessation of treatment. Analyses of cellular scratch assays and the MC903 mouse model of AD suggested that production of sphingolipids by R. mucosa, cholinergic signaling, and flagellin expression may have contributed to therapeutic impact through induction of a TNFR2-mediated epithelial-to-mesenchymal transition. These results suggest that a randomized, placebo-controlled trial of R. mucosa treatment in individuals with AD is warranted and implicate commensals in the maintenance of the skin epithelial barrier.


Subject(s)
Dermatitis, Atopic , Eczema , Methylobacteriaceae , Adult , Child , Dermatitis, Atopic/drug therapy , Humans , Lipids , Skin
4.
PLoS One ; 13(9): e0198862, 2018.
Article in English | MEDLINE | ID: mdl-30208113

ABSTRACT

Keratinocytes are the most abundant cell type in the epidermis. They prevent desiccation and provide immunological and barrier defense against potential pathogens such as Staphylococcus aureus and Candida albicans. The study of this first line of immune defense may be hindered by invasive isolation methods and/or improper culture conditions to support stem cell maintenance and other potential mechanisms contributing to long-term subcultivation in vitro. Primary keratinocytes have been successfully isolated from blister roofs induced by negative pressure, which separates the epidermis from the dermis in vivo in human subjects. This method allows collection of pure epidermal cells without dermal contamination in a minimally invasive manner. However, the isolated keratinocytes differentiate and senesce when cultured in vitro beyond five passages. Here, we present evidence that the Rho kinase (ROCK) inhibitor Y-27632 can be used to effectively increase the proliferative capabilities of keratinocytes isolated using the suction blister method, similar to what has been previously reported for primary keratinocytes isolated using alternative methods. We show that the increase in passage number is directly correlated to delayed differentiation, and that cells passaged long term with the inhibitor retain their ability to stratify in organotypic raft cultures and respond to cytokine treatment; additionally, the late passage cells have a heterogeneous mix of differentiated and non-differentiated cells which may be predicted by a ratio of select differentiation markers. The described method presents a minimally invasive procedure for keratinocyte isolation and prolonged culture that allows analysis of keratinocyte function in both healthy volunteers and patients with dermatologic diseases.


Subject(s)
Amides/pharmacology , Blister/metabolism , Cell Culture Techniques/methods , Epidermis/metabolism , Keratinocytes/metabolism , Pyridines/pharmacology , rho-Associated Kinases/antagonists & inhibitors , Blister/pathology , Cell Proliferation/drug effects , Cells, Cultured , Epidermis/pathology , Humans , Keratinocytes/pathology
5.
FEMS Microbiol Lett ; 365(18)2018 09 01.
Article in English | MEDLINE | ID: mdl-30084923

ABSTRACT

Spores of Bacillus subtilis are encased in a protein coat composed of ∼80 different proteins. Recently, we reconstituted the basement layer of the coat, composed of two structural proteins (SpoVM and SpoIVA) around spore-sized silica beads encased in a lipid bilayer, to create synthetic spore-like particles termed 'SSHELs'. We demonstrated that SSHELs could display thousands of copies of proteins and small molecules of interest covalently linked to SpoIVA. In this study, we investigated the efficacy of SSHELs in delivering vaccines. We show that intramuscular vaccination of mice with undecorated one micron-diameter SSHELs elicited an antibody response against SpoIVA. We further demonstrate that SSHELs covalently modified with a catalytically inactivated staphylococcal alpha toxin variant (HlaH35L), without an adjuvant, resulted in improved protection against Staphylococcus aureus infection in a bacteremia model as compared to vaccination with the antigen alone. Although vaccination with either HlaH35L or HlaH35L conjugated to SSHELs similarly elicited the production of neutralizing antibodies to Hla, we found that a subset of memory T cells was differentially activated when the antigen was delivered on SSHELs. We propose that the particulate nature of SSHELs elicits a more robust immune response to the vaccine that results in superior protection against subsequent S. aureus infection.


Subject(s)
Bacterial Toxins/immunology , Drug Carriers/administration & dosage , Hemolysin Proteins/immunology , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Neutralizing/blood , Bacteremia/prevention & control , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Disease Models, Animal , Hemolysin Proteins/genetics , Injections, Intramuscular , Mice , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Staphylococcal Vaccines/administration & dosage , Staphylococcal Vaccines/genetics , T-Lymphocyte Subsets/immunology , Treatment Outcome , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
6.
J Clin Invest ; 128(8): 3595-3604, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30035749

ABSTRACT

Autosomal dominant hyper IgE syndrome (AD-HIES), or Job's syndrome, is a primary immune deficiency caused by dominant-negative mutations in STAT3. Recurrent Staphylococcus aureus skin abscesses are a defining feature of this syndrome. A widely held hypothesis that defects in peripheral Th17 differentiation confer this susceptibility has never been directly evaluated. To assess the cutaneous immune response in AD-HIES, we induced suction blisters in healthy volunteers (HVs) and patients with AD-HIES and then challenged the wound with lethally irradiated bacteria. We show that cutaneous production of IL-17A and IL-17F was normal in patients with AD-HIES. Overproduction of TNF-α differentiated the responses in AD-HIES from HVs. This was associated with reduced IL-10 family signaling in blister-infiltrating cells and defective epithelial cell function. Mouse models of AD-HIES recapitulated these aberrant epithelial responses to S. aureus and involved defective epithelial-to-mesenchymal transition (EMT) rather than a failure of bacterial killing. Defective responses in mouse models of AD-HIES and primary keratinocyte cultures from patients with AD-HIES could be reversed by TNF-α blockade and by drugs with reported modulatory effects on EMT. Our results identify these as potential therapeutic approaches in patients with AD-HIES suffering S. aureus infections.


Subject(s)
Epithelial Cells/immunology , Furunculosis/immunology , Job Syndrome/immunology , Keratinocytes/immunology , Staphylococcus aureus/immunology , Tumor Necrosis Factor-alpha/immunology , Adult , Animals , Disease Models, Animal , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/immunology , Female , Furunculosis/genetics , Furunculosis/pathology , Humans , Interleukin-17/genetics , Interleukin-17/immunology , Job Syndrome/genetics , Job Syndrome/pathology , Keratinocytes/pathology , Male , Mice , Mice, Transgenic , Tumor Necrosis Factor-alpha/genetics
7.
JCI Insight ; 3(9)2018 05 03.
Article in English | MEDLINE | ID: mdl-29720571

ABSTRACT

The underlying pathology of atopic dermatitis (AD) includes impaired skin barrier function, susceptibility to Staphylococcus aureus skin infection, immune dysregulation, and cutaneous dysbiosis. Our recent investigation into the potential role of Gram-negative skin bacteria in AD revealed that isolates of one particular commensal, Roseomonas mucosa, collected from healthy volunteers (HVs) improved outcomes in mouse and cell culture models of AD. In contrast, isolates of R. mucosa from patients with AD worsened outcomes in these models. These preclinical results suggested that interventions targeting the microbiome could provide therapeutic benefit for patients with AD. As a first test of this hypothesis in humans, 10 adult and 5 pediatric patients were enrolled in an open-label phase I/II safety and activity trial (the Beginning Assessment of Cutaneous Treatment Efficacy for Roseomonas in Atopic Dermatitis trial; BACTERiAD I/II). Treatment with R. mucosa was associated with significant decreases in measures of disease severity, topical steroid requirement, and S. aureus burden. There were no adverse events or treatment complications. We additionally evaluated differentiating bacterial metabolites and topical exposures that may contribute to the skin dysbiosis associated with AD and/or influence future microbiome-based treatments. These early results support continued evaluation of R. mucosa therapy with a placebo-controlled trial.


Subject(s)
Biological Therapy , Dermatitis, Atopic/therapy , Dysbiosis/therapy , Methylobacteriaceae , Microbiota , Skin/microbiology , Adolescent , Adult , Animals , Biological Therapy/adverse effects , Child , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/genetics , Dermatitis, Atopic/microbiology , Dysbiosis/microbiology , Female , Humans , Male , Methylobacteriaceae/isolation & purification , Mice , Severity of Illness Index , Staphylococcus aureus/isolation & purification , Steroids/therapeutic use , Young Adult
8.
J Exp Med ; 215(4): 1079-1090, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29490936

ABSTRACT

Appropriate regulation of IL-17 production in the host can mean the difference between effective control of pathogens and uncontrolled inflammation that causes tissue damage. Investigation of conventional CD4+ T cells (Th17 cells) has yielded invaluable insights into IL-17 function and its regulation. More recently, we and others reported production of IL-17 from innate αß+ T cell populations, which was shown to occur primarily via IL-23R signaling through the transcription factor STAT-3. In our current study, we identify promyelocytic leukemia zinc finger (PLZF)-expressing iNKT, CD4-/CD8+, and CD4-/CD8- (DN) αß+T cells, which produce IL-17 in response to TCR and IL-1 receptor ligation independently of STAT-3 signaling. Notably, this noncanonical pathway of IL-17 production may be important in mucosal defense and is by itself sufficient to control pathogenic Staphylococcus aureus infection at the ocular surface.


Subject(s)
Eye Infections/immunology , Eye Infections/pathology , Immunity, Innate , Interleukin-17/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/metabolism , STAT3 Transcription Factor/metabolism , Animals , Immunologic Memory , Interleukins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mucous Membrane/immunology , Mucous Membrane/microbiology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phosphorylation , Promyelocytic Leukemia Zinc Finger Protein/metabolism , Signal Transduction , Staphylococcus aureus/physiology , T-Lymphocytes/metabolism , Th17 Cells/metabolism , Thymus Gland/metabolism
9.
Article in English | MEDLINE | ID: mdl-30719426

ABSTRACT

Introduction: As therapies for atopic dermatitis (AD) based on live biotherapeutic products (LBP) are developed, the potential displacement of biotherapeutic strains, and species to mucosal sites where they are not naturally found is of investigative interest. However, formal assessment of the toxicity potential of healthy skin commensal organisms has not been reported in the literature. Our previous research indicates that topical application of live Roseomonas mucosa to treat AD was associated with clinical benefit on the skin, but the effects of exposure via inhalation, eye inoculation, and ingestion were unknown. Methods: Herein we report our findings from mice inoculated with commensal strains of R. mucosa, coagulase negative Staphylococci (CNS), and Pseudomonas aeruginosa. Bacterial isolates were collected under clinical trial NCT03018275, however these results do not represent an interventional clinical trial. Results: Our tested R. mucosa isolates did not display significant infection or inflammation. However, neutropenic mice inoculated with CNS had infection without major inflammation in pulmonary models. In contrast, systemic infection generated hepatic and splenic pathology for P. aeruginosa and CNS, which was worsened by the presence of neutropenia. Discussion: Our results suggest that LBP derived from bacteria without significant infectivity histories, such as R. mucosa, may represent safer options than known pathobionts like P. aeruginosa and Staphylococcus spp. Overall, these results suggest that topically applied LBP from select skin commensals are likely to present safe therapeutic options and reinforce our prior clinical findings.


Subject(s)
Bacterial Infections/microbiology , Methylobacteriaceae/growth & development , Probiotics/adverse effects , Pseudomonas aeruginosa/growth & development , Staphylococcus/growth & development , Symbiosis , Virulence , Animals , Bacterial Infections/pathology , Carrier State/microbiology , Disease Models, Animal , Methylobacteriaceae/pathogenicity , Mice , Probiotics/administration & dosage , Pseudomonas aeruginosa/pathogenicity , Staphylococcus/pathogenicity
11.
Sci Immunol ; 2(8)2017 Feb 03.
Article in English | MEDLINE | ID: mdl-28783684

ABSTRACT

IL-22-induced hemopexin promotes nutritional immunity by scavenging iron from Citrobacter rodentium during systemic infection.

12.
Article in English | MEDLINE | ID: mdl-28807923

ABSTRACT

Penicillin binding protein 4 (PBP4) can provide high-level ß-lactam resistance in Staphylococcus aureus A series of missense and promoter mutations associated with pbp4 were detected in strains that displayed high-level resistance. We show here that the missense mutations facilitate the ß-lactam resistance mediated by PBP4 and the promoter mutations lead to overexpression of pbp4 Our results also suggest a cooperative interplay among PBPs for ß-lactam resistance.


Subject(s)
Penicillin-Binding Proteins/genetics , Promoter Regions, Genetic/genetics , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , beta-Lactam Resistance/genetics , Anti-Bacterial Agents/pharmacology , Genome, Bacterial/genetics , Mutation, Missense/genetics , Penicillin-Binding Proteins/biosynthesis , Penicillins/metabolism , Penicillins/pharmacology
13.
Pathogens ; 6(2)2017 Jun 06.
Article in English | MEDLINE | ID: mdl-28587312

ABSTRACT

Autosomal dominant hyper IgE syndrome (AD-HIES) is a primary immunodeficiency caused by a loss-of-function mutation in the Signal Transducer and Activator of Transcription 3 (STAT3). This immune disorder is clinically characterized by increased susceptibility to cutaneous and sinopulmonary infections, in particular with Candida and Staphylococcus aureus. It has recently been recognized that the skin microbiome of patients with AD-HIES is altered with an overrepresentation of certain Gram-negative bacteria and Gram-positive staphylococci. However, these alterations have not been characterized at the species- and strain-level. Since S. aureus infections are influenced by strain-specific expression of virulence factors, information on colonizing strain characteristics may provide insights into host-pathogen interactions and help guide management strategies for treatment and prophylaxis. The aim of this study was to determine whether the immunodeficiency of AD-HIES selects for unique strains of colonizing S. aureus. Using multi-locus sequence typing (MLST), protein A (spa) typing, and PCR-based detection of toxin genes, we performed a detailed analysis of the S. aureus isolates (n = 13) found on the skin of twenty-one patients with AD-HIES. We found a low diversity of sequence types, and an abundance of strains that expressed methicillin resistance, Panton-Valentine leukocidin (PVL), and staphylococcal enterotoxins K and Q (SEK, SEQ). Our results indicate that patients with AD-HIES may often carry antibiotic-resistant strains that harbor key virulence factors.

14.
J Immunol ; 198(11): 4373-4382, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28424238

ABSTRACT

Neutrophils possess multiple antimicrobial mechanisms that are critical for protection of the host against infection with extracellular microbes, such as the bacterial pathogen Staphylococcus aureus Recruitment and activation of neutrophils at sites of infection are driven by cytokine and chemokine signals that directly target neutrophils via specific cell surface receptors. The IL-20 subfamily of cytokines has been reported to act at epithelial sites and contribute to psoriasis, wound healing, and anti-inflammatory effects during S. aureus infection. However, the ability of these cytokines to directly affect neutrophil function remains incompletely understood. In this article, we show that human neutrophils altered their expression of IL-20R chains upon migration and activation in vivo and in vitro. Such activation of neutrophils under conditions mimicking infection with S. aureus conferred responsiveness to IL-20 that manifested as modification of actin polymerization and inhibition of a broad range of actin-dependent functions, including phagocytosis, granule exocytosis, and migration. Consistent with the previously described homeostatic and anti-inflammatory properties of IL-20 on epithelial cells, the current study provides evidence that IL-20 directly targets and inhibits key inflammatory functions of neutrophils during infection with S. aureus.


Subject(s)
Interleukins/metabolism , Neutrophil Activation , Neutrophils/immunology , Neutrophils/physiology , Receptors, Interleukin/immunology , Signal Transduction , Staphylococcus aureus/immunology , Bronchi/cytology , Bronchi/microbiology , Cell Migration Assays, Leukocyte , Cell Movement , Cytokines/biosynthesis , Cytokines/immunology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Exocytosis , Humans , Interleukins/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Phagocytosis , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism
15.
J Infect Dis ; 215(8): 1231-1239, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28329242

ABSTRACT

Multiple candidate vaccines against Staphylococcus aureus infections have failed in clinical trials. Analysis of a recent prematurely halted vaccine trial revealed increased mortality rates among vaccine recipients in whom postsurgical S. aureus infection developed, emphasizing the potential for induction of detrimental immune responses and the need to better understand the requirements for protective immunity against S. aureus. These failures of single-antigen vaccines have prompted ongoing development of multicomponent vaccines to target the multitude of S. aureus virulence factors. In the current study, we used lethally irradiated S. aureus as a model multicomponent vaccine and showed that vaccination of mice decreased survival in a bacteremia challenge model. These deleterious effects were due to a CD4 T-cell-dependent interferon γ response and could be prevented by inhibiting development of this response during vaccination. Our results identify the potential for vaccination to induce pathological immune responses, and they have implications for recent vaccine failures and the design of future staphylococcal vaccines.


Subject(s)
Interferon-gamma/immunology , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/administration & dosage , Staphylococcal Vaccines/adverse effects , Th1 Cells/immunology , Animals , Bacteremia/prevention & control , Female , Methicillin-Resistant Staphylococcus aureus/radiation effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Staphylococcal Infections/immunology
16.
Curr Top Microbiol Immunol ; 409: 419-439, 2017.
Article in English | MEDLINE | ID: mdl-26919865

ABSTRACT

A complex interplay between host and bacterial factors allows Staphylococcus aureus to occupy its niche as a human commensal and a major human pathogen. The role of neutrophils as a critical component of the innate immune response against S. aureus, particularly for control of systemic infection, has been established in both animal models and in humans with acquired and congenital neutrophil dysfunction. The role of the adaptive immune system is less clear. Although deficiencies in adaptive immunity do not result in the marked susceptibility to S. aureus infection that neutrophil dysfunction imparts, emerging evidence suggests both T cell- and B cell-mediated adaptive immunity can influence host susceptibility and control of S. aureus. The contribution of adaptive immunity depends on the context and site of infection and can be either beneficial or detrimental to the host. Furthermore, S. aureus has evolved mechanisms to manipulate adaptive immune responses to its advantage. In this chapter, we will review the evidence for the role of adaptive immunity during S. aureus infections. Further elucidation of this role will be important to understand how it influences susceptibility to infection and to appropriately design vaccines that elicit adaptive immune responses to protect against subsequent infections.


Subject(s)
Staphylococcus aureus , Adaptive Immunity , Animals , Humans , Immunity, Innate , Staphylococcal Infections
17.
J Leukoc Biol ; 101(1): 307-320, 2017 01.
Article in English | MEDLINE | ID: mdl-27515950

ABSTRACT

Cellular lysates from PPD+ donors have been reported to transfer tuberculin reactivity to naïve recipients, but not diphtheria reactivity, and vice versa. A historically controversial topic, the terms "transfer factor" and "DLE" were used to characterize the reactivity-transferring properties of lysates. Intrigued by these reported phenomena, we found that the cellular extract derived from antigen-specific memory CD8+ T cells induces IL-6 from antigen-matched APCs. This ultimately elicits IL-17 from bystander memory CD8+ T cells. We have identified that dialyzable peptide sequences, S100a9, and the TCR ß chain from CD8+ T cells contribute to the molecular nature of this activity. We further show that extracts from antigen-targeted T cells enhance immunity to Staphylococcus aureus and Candida albicans These effects are sensitive to immunization protocols and extraction methodology in ways that may explain past discrepancies in the reproducibility of passive cellular immunity.


Subject(s)
Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dialysis , Animals , Epitopes/immunology , Humans , Immunity , Immunologic Memory , Interleukin-17/metabolism , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Biological , Receptors, Antigen, T-Cell/metabolism , S100 Proteins/metabolism , Spleen/pathology
18.
JCI Insight ; 1(10)2016 Jul 07.
Article in English | MEDLINE | ID: mdl-27478874

ABSTRACT

Atopic dermatitis (AD) is characterized by reduced barrier function, reduced innate immune activation, and susceptibility to Staphylococcus aureus. Host susceptibility factors are suggested by monogenic disorders associated with AD-like phenotypes and can be medically modulated. S. aureus contributes to AD pathogenesis and can be mitigated by antibiotics and bleach baths. Recent work has revealed that the skin microbiome differs significantly between healthy controls and patients with AD, including decreased Gram-negative bacteria in AD. However, little is known about the potential therapeutic benefit of microbiome modulation. To evaluate whether parameters of AD pathogenesis are altered after exposure to different culturable Gram-negative bacteria (CGN) collected from human skin, CGN were collected from healthy controls and patients with AD. Then, effects on cellular and culture-based models of immune, epithelial, and bacterial function were evaluated. Representative strains were evaluated in the MC903 mouse model of AD. We found that CGN taken from healthy volunteers but not from patients with AD were associated with enhanced barrier function, innate immunity activation, and control of S. aureus. Treatment with CGN from healthy controls improved outcomes in a mouse model of AD. These findings suggest that a live-biotherapeutic approach may hold promise for treatment of patients with AD.

19.
BMC Microbiol ; 16: 60, 2016 Apr 06.
Article in English | MEDLINE | ID: mdl-27052736

ABSTRACT

BACKGROUND: Commensal Gram-negative (CGN) microbiota have been identified on human skin by DNA sequencing; however, methods to reliably culture viable Gram-negative skin organisms have not been previously described. RESULTS: Through the use of selective antibiotics and minimal media we developed methods to culture CGN from skin swabs. We identified several previously uncharacterized CGN at the species level by optimizing growth conditions and limiting the inhibitory effects of nutrient shock, temperature, and bacterial competition, factors that may have previously limited CGN isolation from skin cultures. CONCLUSIONS: Our protocol will permit future functional studies on the influences of CGN on skin homeostasis and disease.


Subject(s)
Bacteriological Techniques/methods , Gram-Negative Bacteria/growth & development , Skin/microbiology , Culture Media , Gram-Negative Bacteria/isolation & purification , Humans , Microbiota
20.
PLoS Pathog ; 11(12): e1005293, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26679537

ABSTRACT

Candida is the most common human fungal pathogen and causes systemic infections that require neutrophils for effective host defense. Humans deficient in the C-type lectin pathway adaptor protein CARD9 develop spontaneous fungal disease that targets the central nervous system (CNS). However, how CARD9 promotes protective antifungal immunity in the CNS remains unclear. Here, we show that a patient with CARD9 deficiency had impaired neutrophil accumulation and induction of neutrophil-recruiting CXC chemokines in the cerebrospinal fluid despite uncontrolled CNS Candida infection. We phenocopied the human susceptibility in Card9-/- mice, which develop uncontrolled brain candidiasis with diminished neutrophil accumulation. The induction of neutrophil-recruiting CXC chemokines is significantly impaired in infected Card9-/- brains, from both myeloid and resident glial cellular sources, whereas cell-intrinsic neutrophil chemotaxis is Card9-independent. Taken together, our data highlight the critical role of CARD9-dependent neutrophil trafficking into the CNS and provide novel insight into the CNS fungal susceptibility of CARD9-deficient humans.


Subject(s)
CARD Signaling Adaptor Proteins/immunology , Candidiasis/immunology , Central Nervous System Infections/immunology , Immunologic Deficiency Syndromes/immunology , Neutrophil Infiltration/immunology , Animals , Blotting, Western , CARD Signaling Adaptor Proteins/deficiency , Female , Flow Cytometry , Humans , Immunologic Deficiency Syndromes/microbiology , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...