Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 184(3): 615-627.e17, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33453153

ABSTRACT

The microbiota shields the host against infections in a process known as colonization resistance. How infections themselves shape this fundamental process remains largely unknown. Here, we show that gut microbiota from previously infected hosts display enhanced resistance to infection. This long-term functional remodeling is associated with altered bile acid metabolism leading to the expansion of taxa that utilize the sulfonic acid taurine. Notably, supplying exogenous taurine alone is sufficient to induce this alteration in microbiota function and enhance resistance. Mechanistically, taurine potentiates the microbiota's production of sulfide, an inhibitor of cellular respiration, which is key to host invasion by numerous pathogens. As such, pharmaceutical sequestration of sulfide perturbs the microbiota's composition and promotes pathogen invasion. Together, this work reveals a process by which the host, triggered by infection, can deploy taurine as a nutrient to nourish and train the microbiota, promoting its resistance to subsequent infection.


Subject(s)
Gastrointestinal Microbiome , Host-Pathogen Interactions , Animals , Bacterial Infections/immunology , Bacterial Infections/microbiology , Colony Count, Microbial , Gastrointestinal Microbiome/drug effects , Host-Pathogen Interactions/drug effects , Immunity , Mice, Inbred C57BL , Sulfides/metabolism , Taurine/pharmacology
2.
Cell ; 184(12): 3075-3079, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34115967

ABSTRACT

NIH has acknowledged and committed to ending structural racism. The framework for NIH's approach, summarized here, includes understanding barriers; developing robust health disparities/equity research; improving its internal culture; being transparent and accountable; and changing the extramural ecosystem so that diversity, equity, and inclusion are reflected in funded research and the biomedical workforce.


Subject(s)
Biomedical Research , National Institutes of Health (U.S.) , Systemic Racism , Cultural Diversity , Humans , Research Support as Topic/economics , United States
3.
Cell ; 169(3): 378-380, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28431240

ABSTRACT

Designing successful microbiota-based therapies requires in-depth understanding of the ecological foundations of this community. In this issue, two studies by Whitaker et al. and Lim et al. provide refined genetic tools for dissecting the spatial organization and temporal dynamics of bacterial communities at the single-cell and -gene levels.


Subject(s)
Microbiota
4.
Cell ; 164(6): 1288-1300, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26967294

ABSTRACT

Human-associated microbiota form and stabilize communities based on interspecies interactions. We review how these microbe-microbe and microbe-host interactions are communicated to shape communities over a human's lifespan, including periods of health and disease. Modeling and dissecting signaling in host-associated communities is crucial to understand their function and will open the door to therapies that prevent or correct microbial community dysfunction to promote health and treat disease.


Subject(s)
Microbiota , Signal Transduction , Animals , Bacteria/classification , Bacteria/metabolism , Humans , Microbial Interactions
5.
Cell ; 165(4): 854-66, 2016 May 05.
Article in English | MEDLINE | ID: mdl-27153496

ABSTRACT

Biogeography and individuality shape the structural and functional composition of the human skin microbiome. To explore these factors' contribution to skin microbial community stability, we generated metagenomic sequence data from longitudinal samples collected over months and years. Analyzing these samples using a multi-kingdom, reference-based approach, we found that despite the skin's exposure to the external environment, its bacterial, fungal, and viral communities were largely stable over time. Site, individuality, and phylogeny were all determinants of stability. Foot sites exhibited the most variability; individuals differed in stability; and transience was a particular characteristic of eukaryotic viruses, which showed little site-specificity in colonization. Strain and single-nucleotide variant-level analysis showed that individuals maintain, rather than reacquire, prevalent microbes from the environment. Longitudinal stability of skin microbial communities generates hypotheses about colonization resistance and empowers clinical studies exploring alterations observed in disease states.


Subject(s)
Bacteria/classification , Fungi/classification , Microbiota , Skin/microbiology , Viruses/classification , Bacteria/isolation & purification , Bacterial Physiological Phenomena , DNA Viruses/isolation & purification , Fungi/isolation & purification , Fungi/physiology , Homeostasis , Humans , Propionibacterium acnes/isolation & purification , Skin Physiological Phenomena , Symbiosis , Virus Physiological Phenomena , Viruses/isolation & purification
6.
Immunity ; 54(10): 2321-2337.e10, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34582748

ABSTRACT

Hair follicles (HFs) function as hubs for stem cells, immune cells, and commensal microbes, which must be tightly regulated during homeostasis and transient inflammation. Here we found that transmembrane endopeptidase ADAM10 expression in upper HFs was crucial for regulating the skin microbiota and protecting HFs and their stem cell niche from inflammatory destruction. Ablation of the ADAM10-Notch signaling axis impaired the innate epithelial barrier and enabled Corynebacterium species to predominate the microbiome. Dysbiosis triggered group 2 innate lymphoid cell-mediated inflammation in an interleukin-7 (IL-7) receptor-, S1P receptor 1-, and CCR6-dependent manner, leading to pyroptotic cell death of HFs and irreversible alopecia. Double-stranded RNA-induced ablation models indicated that the ADAM10-Notch signaling axis bolsters epithelial innate immunity by promoting ß-defensin-6 expression downstream of type I interferon responses. Thus, ADAM10-Notch signaling axis-mediated regulation of host-microbial symbiosis crucially protects HFs from inflammatory destruction, which has implications for strategies to sustain tissue integrity during chronic inflammation.


Subject(s)
ADAM10 Protein/immunology , Amyloid Precursor Protein Secretases/immunology , Dysbiosis/immunology , Hair Follicle/pathology , Lymphocytes/immunology , Membrane Proteins/immunology , Receptors, Notch/immunology , Skin/microbiology , Alopecia/immunology , Alopecia/pathology , Animals , Corynebacterium , Dysbiosis/pathology , Female , Hair Follicle/immunology , Immunity, Innate , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Mice , Signal Transduction/immunology , Skin/immunology , Skin/pathology
7.
Proc Natl Acad Sci U S A ; 120(47): e2310585120, 2023 Nov 21.
Article in English | MEDLINE | ID: mdl-37956283

ABSTRACT

Human skin is stably colonized by a distinct microbiota that functions together with epidermal cells to maintain a protective physical barrier. Staphylococcus, a prominent genus of the skin microbiota, participates in colonization resistance, tissue repair, and host immune regulation in strain-specific manners. To unlock the potential of engineering skin microbial communities, we aim to characterize the diversity of this genus within the context of the skin environment. We reanalyzed an extant 16S rRNA amplicon dataset obtained from distinct body sites of healthy volunteers, providing a detailed biogeographic depiction of staphylococcal species that colonize our skin. S. epidermidis, S. capitis, and S. hominis were the most abundant staphylococcal species present in all volunteers and were detected at all body sites. Pan-genome analysis of isolates from these three species revealed that the genus-core was dominated by central metabolism genes. Species-restricted-core genes encoded known host colonization functions. The majority (~68%) of genes were detected only in a fraction of isolate genomes, underscoring the immense strain-specific gene diversity. Conspecific genomes grouped into phylogenetic clades, exhibiting body site preference. Each clade was enriched for distinct gene sets that are potentially involved in site tropism. Finally, we conducted gene expression studies of select isolates showing variable growth phenotypes in skin-like medium. In vitro expression revealed extensive intra- and inter-species gene expression variation, substantially expanding the functional diversification within each species. Our study provides an important resource for future ecological and translational studies to examine the role of shared and strain-specific staphylococcal genes within the skin environment.


Subject(s)
Skin , Staphylococcus , Humans , Staphylococcus/genetics , Phylogeny , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , Staphylococcus epidermidis/genetics , Genomics
8.
N Engl J Med ; 385(10): 921-929, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34469647

ABSTRACT

Human papillomavirus (HPV) infections underlie a wide spectrum of both benign and malignant epithelial diseases. In this report, we describe the case of a young man who had encephalitis caused by herpes simplex virus during adolescence and currently presented with multiple recurrent skin and mucosal lesions caused by HPV. The patient was found to have a pathogenic germline mutation in the X-linked interleukin-2 receptor subunit gamma gene (IL2RG), which was somatically reverted in T cells but not in natural killer (NK) cells. Allogeneic hematopoietic-cell transplantation led to restoration of NK cytotoxicity, with normalization of the skin microbiome and persistent remission of all HPV-related diseases. NK cytotoxicity appears to play a role in containing HPV colonization and the ensuing HPV-related hyperplastic or dysplastic lesions. (Funded by the National Institutes of Health and the Herbert Irving Comprehensive Cancer Center Flow Cytometry Shared Resources.).


Subject(s)
Germ-Line Mutation , Hematopoietic Stem Cell Transplantation , Killer Cells, Natural/physiology , Papillomavirus Infections/therapy , Cytotoxicity, Immunologic , Encephalitis/virology , Female , Humans , Killer Cells, Natural/drug effects , Male , Microbiota/drug effects , Natural Killer T-Cells/physiology , Papillomaviridae , Papillomavirus Infections/genetics , Papillomavirus Infections/immunology , Pedigree , Skin/microbiology , Transplantation, Homologous , Young Adult
9.
Immunity ; 43(5): 842-4, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26588777

ABSTRACT

How immune tolerance is maintained in the skin remains unclear. In this issue of Immunity, Rosenblum and colleagues demonstrate that tolerance to commensal bacteria is established during the neonatal period via regulatory T cells. Defining the crucial window during which commensal-specific tolerance is achieved has strategic implications for the induction of tolerance in allergic diseases.


Subject(s)
Animals, Newborn/immunology , Skin/immunology , Skin/microbiology , Staphylococcal Infections/immunology , Staphylococcus epidermidis/immunology , T-Lymphocytes, Regulatory/immunology , Animals
10.
N Engl J Med ; 379(26): 2529-2539, 2018 12 27.
Article in English | MEDLINE | ID: mdl-30586509

ABSTRACT

BACKGROUND: Plumbing systems are an infrequent but known reservoir for opportunistic microbial pathogens that can infect hospitalized patients. In 2016, a cluster of clinical sphingomonas infections prompted an investigation. METHODS: We performed whole-genome DNA sequencing on clinical isolates of multidrug-resistant Sphingomonas koreensis identified from 2006 through 2016 at the National Institutes of Health (NIH) Clinical Center. We cultured S. koreensis from the sinks in patient rooms and performed both whole-genome and shotgun metagenomic sequencing to identify a reservoir within the infrastructure of the hospital. These isolates were compared with clinical and environmental S. koreensis isolates obtained from other institutions. RESULTS: The investigation showed that two isolates of S. koreensis obtained from the six patients identified in the 2016 cluster were unrelated, but four isolates shared more than 99.92% genetic similarity and were resistant to multiple antibiotic agents. Retrospective analysis of banked clinical isolates of sphingomonas from the NIH Clinical Center revealed the intermittent recovery of a clonal strain over the past decade. Unique single-nucleotide variants identified in strains of S. koreensis elucidated the existence of a reservoir in the hospital plumbing. Clinical S. koreensis isolates from other facilities were genetically distinct from the NIH isolates. Hospital remediation strategies were guided by results of microbiologic culturing and fine-scale genomic analyses. CONCLUSIONS: This genomic and epidemiologic investigation suggests that S. koreensis is an opportunistic human pathogen that both persisted in the NIH Clinical Center infrastructure across time and space and caused health care-associated infections. (Funded by the NIH Intramural Research Programs.).


Subject(s)
Cross Infection/microbiology , Disease Reservoirs/microbiology , Gram-Negative Bacterial Infections/microbiology , Sanitary Engineering , Sphingomonas/genetics , Anti-Bacterial Agents/pharmacology , Hospitals, Federal , Humans , Metagenomics , Microbial Sensitivity Tests , National Institutes of Health (U.S.) , Retrospective Studies , Sphingomonas/drug effects , Sphingomonas/isolation & purification , United States , Water Supply , Whole Genome Sequencing
11.
Nature ; 520(7545): 104-8, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25539086

ABSTRACT

The skin represents the primary interface between the host and the environment. This organ is also home to trillions of microorganisms that play an important role in tissue homeostasis and local immunity. Skin microbial communities are highly diverse and can be remodelled over time or in response to environmental challenges. How, in the context of this complexity, individual commensal microorganisms may differentially modulate skin immunity and the consequences of these responses for tissue physiology remains unclear. Here we show that defined commensals dominantly affect skin immunity and identify the cellular mediators involved in this specification. In particular, colonization with Staphylococcus epidermidis induces IL-17A(+) CD8(+) T cells that home to the epidermis, enhance innate barrier immunity and limit pathogen invasion. Commensal-specific T-cell responses result from the coordinated action of skin-resident dendritic cell subsets and are not associated with inflammation, revealing that tissue-resident cells are poised to sense and respond to alterations in microbial communities. This interaction may represent an evolutionary means by which the skin immune system uses fluctuating commensal signals to calibrate barrier immunity and provide heterologous protection against invasive pathogens. These findings reveal that the skin immune landscape is a highly dynamic environment that can be rapidly and specifically remodelled by encounters with defined commensals, findings that have profound implications for our understanding of tissue-specific immunity and pathologies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Skin/immunology , Skin/microbiology , Symbiosis/immunology , Animals , Antigens, Bacterial/immunology , CD8-Positive T-Lymphocytes/cytology , Dendritic Cells/cytology , Humans , Immunity, Innate/immunology , Interleukin-17/immunology , Langerhans Cells/cytology , Langerhans Cells/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Primates , Skin/cytology , Staphylococcus epidermidis/immunology
12.
Nature ; 514(7520): 59-64, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25279917

ABSTRACT

The varied topography of human skin offers a unique opportunity to study how the body's microenvironments influence the functional and taxonomic composition of microbial communities. Phylogenetic marker gene-based studies have identified many bacteria and fungi that colonize distinct skin niches. Here metagenomic analyses of diverse body sites in healthy humans demonstrate that local biogeography and strong individuality define the skin microbiome. We developed a relational analysis of bacterial, fungal and viral communities, which showed not only site specificity but also individual signatures. We further identified strain-level variation of dominant species as heterogeneous and multiphyletic. Reference-free analyses captured the uncharacterized metagenome through the development of a multi-kingdom gene catalogue, which was used to uncover genetic signatures of species lacking reference genomes. This work is foundational for human disease studies investigating inter-kingdom interactions, metabolic changes and strain tracking, and defines the dual influence of biogeography and individuality on microbial composition and function.


Subject(s)
Metagenome , Skin/microbiology , Skin/virology , Bacteriophages/genetics , Bacteriophages/isolation & purification , Female , Genome, Bacterial/genetics , Genome, Fungal/genetics , Genome, Viral/genetics , Genomics , Healthy Volunteers , Humans , Male , Metagenome/genetics , Phylogeny , Propionibacterium acnes/genetics , Propionibacterium acnes/isolation & purification , Propionibacterium acnes/virology , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/isolation & purification , Staphylococcus epidermidis/virology , Symbiosis
13.
J Allergy Clin Immunol ; 143(4): 1426-1443.e6, 2019 04.
Article in English | MEDLINE | ID: mdl-30240702

ABSTRACT

BACKGROUND: Atopic dermatitis (AD) is associated with epidermal barrier defects, dysbiosis, and skin injury caused by scratching. In particular, the barrier-defective epidermis in patients with AD with loss-of-function filaggrin mutations has increased IL-1α and IL-1ß levels, but the mechanisms by which IL-1α, IL-1ß, or both are induced and whether they contribute to the aberrant skin inflammation in patients with AD is unknown. OBJECTIVE: We sought to determine the mechanisms through which skin injury, dysbiosis, and increased epidermal IL-1α and IL-1ß levels contribute to development of skin inflammation in a mouse model of injury-induced skin inflammation in filaggrin-deficient mice without the matted mutation (ft/ft mice). METHODS: Skin injury of wild-type, ft/ft, and myeloid differentiation primary response gene-88-deficient ft/ft mice was performed, and ensuing skin inflammation was evaluated by using digital photography, histologic analysis, and flow cytometry. IL-1α and IL-1ß protein expression was measured by means of ELISA and visualized by using immunofluorescence and immunoelectron microscopy. Composition of the skin microbiome was determined by using 16S rDNA sequencing. RESULTS: Skin injury of ft/ft mice induced chronic skin inflammation involving dysbiosis-driven intracellular IL-1α release from keratinocytes. IL-1α was necessary and sufficient for skin inflammation in vivo and secreted from keratinocytes by various stimuli in vitro. Topical antibiotics or cohousing of ft/ft mice with unaffected wild-type mice to alter or intermix skin microbiota, respectively, resolved the skin inflammation and restored keratinocyte intracellular IL-1α localization. CONCLUSIONS: Taken together, skin injury, dysbiosis, and filaggrin deficiency triggered keratinocyte intracellular IL-1α release that was sufficient to drive chronic skin inflammation, which has implications for AD pathogenesis and potential therapeutic targets.


Subject(s)
Dermatitis, Atopic/metabolism , Inflammation/metabolism , Interleukin-1alpha/metabolism , Intermediate Filament Proteins/deficiency , Keratinocytes/metabolism , Animals , Dermatitis, Atopic/immunology , Dermatitis, Atopic/microbiology , Dysbiosis/immunology , Dysbiosis/metabolism , Filaggrin Proteins , Inflammation/immunology , Inflammation/microbiology , Interleukin-1alpha/immunology , Keratinocytes/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout
14.
Nature ; 498(7454): 367-70, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-23698366

ABSTRACT

Traditional culture-based methods have incompletely defined the microbial landscape of common recalcitrant human fungal skin diseases, including athlete's foot and toenail infections. Skin protects humans from invasion by pathogenic microorganisms and provides a home for diverse commensal microbiota. Bacterial genomic sequence data have generated novel hypotheses about species and community structures underlying human disorders. However, microbial diversity is not limited to bacteria; microorganisms such as fungi also have major roles in microbial community stability, human health and disease. Genomic methodologies to identify fungal species and communities have been limited compared with those that are available for bacteria. Fungal evolution can be reconstructed with phylogenetic markers, including ribosomal RNA gene regions and other highly conserved genes. Here we sequenced and analysed fungal communities of 14 skin sites in 10 healthy adults. Eleven core-body and arm sites were dominated by fungi of the genus Malassezia, with only species-level classifications revealing fungal-community composition differences between sites. By contrast, three foot sites--plantar heel, toenail and toe web--showed high fungal diversity. Concurrent analysis of bacterial and fungal communities demonstrated that physiologic attributes and topography of skin differentially shape these two microbial communities. These results provide a framework for future investigation of the contribution of interactions between pathogenic and commensal fungal and bacterial communities to the maintainenace of human health and to disease pathogenesis.


Subject(s)
Bacteria/isolation & purification , Biodiversity , Fungi/isolation & purification , Skin/microbiology , Adult , Bacteria/classification , Bacteria/genetics , Databases, Genetic , District of Columbia , Female , Fungi/classification , Fungi/genetics , Health , Homeostasis , Humans , Malassezia/classification , Malassezia/genetics , Malassezia/isolation & purification , Male , Molecular Sequence Data , Skin/anatomy & histology , Young Adult
15.
J Allergy Clin Immunol ; 139(1): 166-172, 2017 01.
Article in English | MEDLINE | ID: mdl-27609659

ABSTRACT

BACKGROUND: Disease flares of established atopic dermatitis (AD) are generally associated with a low-diversity skin microbiota and Staphylococcus aureus dominance. The temporal transition of the skin microbiome between early infancy and the dysbiosis of established AD is unknown. METHODS: We randomly selected 50 children from the Cork Babies After SCOPE: Evaluating the Longitudinal Impact Using Neurological and Nutritional Endpoints (BASELINE) longitudinal birth cohort for microbiome sampling at 3 points in the first 6 months of life at 4 skin sites relevant to AD: the antecubital and popliteal fossae, nasal tip, and cheek. We identified 10 infants with AD and compared them with 10 randomly selected control infants with no AD. We performed bacterial 16S ribosomal RNA sequencing and analysis directly from clinical samples. RESULTS: Bacterial community structures and diversity shifted over time, suggesting that age strongly affects the skin microbiome in infants. Unlike established AD, these patients with infantile AD did not have noticeably dysbiotic communities before or with disease and were not colonized by S aureus. In comparing patients and control subjects, infants who had affected skin at month 12 had statistically significant differences in bacterial communities on the antecubital fossa at month 2 compared with infants who were unaffected at month 12. In particular, commensal staphylococci were significantly less abundant in infants affected at month 12, suggesting that this genus might protect against the later development of AD. CONCLUSIONS: This study suggests that 12-month-old infants with AD were not colonized with S aureus before having AD. Additional studies are needed to confirm whether colonization with commensal staphylococci modulates skin immunity and attenuates development of AD.


Subject(s)
Dermatitis, Atopic/microbiology , Microbiota , Skin/microbiology , Bacteria/genetics , Child, Preschool , Female , Filaggrin Proteins , Humans , Infant , Intermediate Filament Proteins/genetics , Male , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , Risk Factors
16.
Genome Res ; 23(7): 1155-62, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23564252

ABSTRACT

Bacterial whole-genome sequencing (WGS) of human pathogens has provided unprecedented insights into the evolution of antibiotic resistance. Most studies have focused on identification of resistance mutations, leaving one to speculate on the fate of these mutants once the antibiotic selective pressure is removed. We performed WGS on longitudinal isolates of Acinetobacter baumannii from patients undergoing colistin treatment, and upon subsequent drug withdrawal. In each of the four patients, colistin resistance evolved via mutations at the pmr locus. Upon colistin withdrawal, an ancestral susceptible strain outcompeted resistant isolates in three of the four cases. In the final case, resistance was also lost, but by a compensatory inactivating mutation in the transcriptional regulator of the pmr locus. Notably, this inactivating mutation reduced the probability of reacquiring colistin resistance when subsequently challenged in vitro. On face value, these results supported an in vivo fitness cost preventing the evolution of stable colistin resistance. However, more careful analysis of WGS data identified genomic evidence for stable colistin resistance undetected by clinical microbiological assays. Transcriptional studies validated this genomic hypothesis, showing increased pmr expression of the initial isolate. Moreover, altering the environmental growth conditions of the clinical assay recapitulated the classification as colistin resistant. Additional targeted sequencing revealed that this isolate evolved undetected in a patient undergoing colistin treatment, and was then transmitted to other hospitalized patients, further demonstrating its stability in the absence of colistin. This study provides a unique window into mutational pathways taken in response to antibiotic pressure in vivo, and demonstrates the potential for genome sequence data to predict resistance phenotypes.


Subject(s)
Acinetobacter baumannii/drug effects , Acinetobacter baumannii/genetics , Anti-Bacterial Agents/pharmacology , Colistin/pharmacology , Drug Resistance, Bacterial/genetics , Genome, Bacterial , Genomics , Acinetobacter Infections/drug therapy , Anti-Bacterial Agents/therapeutic use , Colistin/therapeutic use , Gene Expression Regulation, Bacterial , Genes, Bacterial , Genetic Fitness , High-Throughput Nucleotide Sequencing , Humans , Microbial Sensitivity Tests , Mutation , Polymorphism, Single Nucleotide
17.
Genome Res ; 23(12): 2103-14, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24170601

ABSTRACT

While landmark studies have shown that microbiota activate and educate host immunity, how immune systems shape microbiomes and contribute to disease is incompletely characterized. Primary immunodeficiency (PID) patients suffer recurrent microbial infections, providing a unique opportunity to address this issue. To investigate the potential influence of host immunity on the skin microbiome, we examined skin microbiomes in patients with rare monogenic PIDs: hyper-IgE (STAT3-deficient), Wiskott-Aldrich, and dedicator of cytokinesis 8 syndromes. While specific immunologic defects differ, a shared hallmark is atopic dermatitis (AD)-like eczema. We compared bacterial and fungal skin microbiomes (41 PID, 13 AD, 49 healthy controls) at four clinically relevant sites representing the major skin microenvironments. PID skin displayed increased ecological permissiveness with altered population structures, decreased site specificity and temporal stability, and colonization with microbial species not observed in controls, including Clostridium species and Serratia marcescens. Elevated fungal diversity and increased representation of opportunistic fungi (Candida, Aspergillus) supported increased PID skin permissiveness, suggesting that skin may serve as a reservoir for the recurrent fungal infections observed in these patients. The overarching theme of increased ecological permissiveness in PID skin was counterbalanced by the maintenance of a phylum barrier in which colonization remained restricted to typical human-associated phyla. Clinical parameters, including markers of disease severity, were positively correlated with prevalence of Staphylococcus, Corynebacterium, and other less abundant taxa. This study examines differences in microbial colonization and community stability in PID skin and informs our understanding of host-microbiome interactions, suggesting a bidirectional dialogue between skin commensals and the host organism.


Subject(s)
Bacteria/genetics , Dermatitis, Atopic/microbiology , Fungi/genetics , Immunologic Deficiency Syndromes/microbiology , Microbiota/genetics , Skin/microbiology , Adolescent , Adult , Bacteria/classification , Bacteria/pathogenicity , Child , Child, Preschool , Corynebacterium/genetics , Corynebacterium/immunology , Dermatitis, Atopic/immunology , Female , Fungi/classification , Fungi/pathogenicity , Host-Pathogen Interactions , Humans , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/pathology , Male , Microbiota/immunology , RNA, Ribosomal, 16S/genetics , Skin/immunology , Staphylococcus/genetics , Staphylococcus/immunology , Young Adult
18.
J Clin Microbiol ; 54(4): 1167-70, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26888898

ABSTRACT

Perirectal surveillance cultures and a stool culture grew Aeromonas species from three patients over a 6-week period and were without epidemiological links. Detection of the blaKPC-2 gene in one isolate prompted inclusion of non-Enterobacteriaceae in our surveillance culture workup. Whole-genome sequencing confirmed that the isolates were unrelated and provided data for Aeromonas reference genomes.


Subject(s)
Aeromonas hydrophila/enzymology , Aeromonas hydrophila/genetics , Anal Canal/microbiology , Bacterial Proteins/genetics , Genome, Bacterial , Sequence Analysis, DNA , beta-Lactamases/genetics , Adult , Aeromonas hydrophila/classification , Aeromonas hydrophila/isolation & purification , Epidemiological Monitoring , Feces/microbiology , Genetic Variation , Genotype , Humans , Molecular Epidemiology
19.
Annu Rev Genomics Hum Genet ; 13: 151-70, 2012.
Article in English | MEDLINE | ID: mdl-22703178

ABSTRACT

The human genome has been referred to as the blueprint of human biology. In this review we consider an essential but largely ignored overlay to that blueprint, the human microbiome, which is composed of those microbes that live in and on our bodies. The human microbiome is a source of genetic diversity, a modifier of disease, an essential component of immunity, and a functional entity that influences metabolism and modulates drug interactions. Characterization and analysis of the human microbiome have been greatly catalyzed by advances in genomic technologies. We discuss how these technologies have shaped this emerging field of study and advanced our understanding of the human microbiome. We also identify future challenges, many of which are common to human genetic studies, and predict that in the future, analyzing genetic variation and risk of human disease will sometimes necessitate the integration of human and microbial genomic data sets.


Subject(s)
Metagenome , Animals , Bacteria/drug effects , Bacteria/genetics , Drug Resistance, Bacterial , Gastrointestinal Tract/microbiology , Humans , Molecular Typing , Mouth/microbiology , Sequence Analysis, DNA , Skin/microbiology , Urogenital System/microbiology
20.
Genome Res ; 22(5): 850-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22310478

ABSTRACT

Atopic dermatitis (AD) has long been associated with Staphylococcus aureus skin colonization or infection and is typically managed with regimens that include antimicrobial therapies. However, the role of microbial communities in the pathogenesis of AD is incompletely characterized. To assess the relationship between skin microbiota and disease progression, 16S ribosomal RNA bacterial gene sequencing was performed on DNA obtained directly from serial skin sampling of children with AD. The composition of bacterial communities was analyzed during AD disease states to identify characteristics associated with AD flares and improvement post-treatment. We found that microbial community structures at sites of disease predilection were dramatically different in AD patients compared with controls. Microbial diversity during AD flares was dependent on the presence or absence of recent AD treatments, with even intermittent treatment linked to greater bacterial diversity than no recent treatment. Treatment-associated changes in skin bacterial diversity suggest that AD treatments diversify skin bacteria preceding improvements in disease activity. In AD, the proportion of Staphylococcus sequences, particularly S. aureus, was greater during disease flares than at baseline or post-treatment, and correlated with worsened disease severity. Representation of the skin commensal S. epidermidis also significantly increased during flares. Increases in Streptococcus, Propionibacterium, and Corynebacterium species were observed following therapy. These findings reveal linkages between microbial communities and inflammatory diseases such as AD, and demonstrate that as compared with culture-based studies, higher resolution examination of microbiota associated with human disease provides novel insights into global shifts of bacteria relevant to disease progression and treatment.


Subject(s)
Dermatitis, Atopic/microbiology , Metagenome , Skin/microbiology , Adolescent , Case-Control Studies , Child , Child, Preschool , Databases, Genetic , Dermatitis, Atopic/pathology , Humans , Molecular Typing , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , Sequence Analysis, RNA , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus/genetics , Statistics, Nonparametric
SELECTION OF CITATIONS
SEARCH DETAIL