Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Zoonoses Public Health ; 69(5): 550-559, 2022 08.
Article in English | MEDLINE | ID: mdl-35420715

ABSTRACT

It has been suggested that pets play a critical role in the maintenance of methicillin-resistant (MR) and multidrug-resistant (MDR) Staphylococcus spp. in the household. We examined risk factors for carriage of antimicrobial-resistant coagulase-positive staphylococci, with particular attention to Staphylococcus aureus and Staphylococcus pseudintermedius isolated from pets living in households of people diagnosed with methicillin-resistant S. aureus (MRSA) skin or soft-tissue infection. We analyzed data collected cross-sectionally from a study conducted in 2012 that evaluated the transmission of MRSA and other staphylococci from humans, their pets and the environment (Pets and Environmental Transmission of Staphylococci [PETS] study). We used unadjusted and adjusted stratified logistic regression analyses with household-clustered standard errors to evaluate the association between demographic, healthcare-related, contact-related and environmental risk factors and MDR Staphylococcus spp. isolated from dogs and cats. Staphylococcal isolates obtained from dogs (n = 63) and cats (n = 47) were included in these analyses. The use of oral or injectable antimicrobials by the pets during the prior year was the main risk factor of interest. Based on our results, 50% (12/24) of S. aureus, 3.3% (1/30) of S. pseudintermedius and 25% (14/56) of other coagulase-positive staphylococci (CPS) were determined to be MDR. S. aureus isolates were more likely to be MDR compared with S. pseudintermedius. We did not find a significant statistical association between the use of oral or injectable antimicrobials in the prior year and the presence of MDR bacteria. The results suggest that drivers of antimicrobial resistance in household staphylococci may vary by bacterial species, which could have implications for one health intervention strategies for staphylococci and inform the investigation of other reverse zoonoses, such as COVID-19.


Subject(s)
Anti-Infective Agents , COVID-19 , Cat Diseases , Dog Diseases , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Animals , Anti-Bacterial Agents/pharmacology , COVID-19/veterinary , Cat Diseases/microbiology , Cats , Coagulase , Dog Diseases/epidemiology , Dog Diseases/microbiology , Dogs , Drug Resistance, Bacterial , Humans , Pets/microbiology , Risk Factors , Staphylococcal Infections/epidemiology , Staphylococcal Infections/microbiology , Staphylococcal Infections/veterinary , Staphylococcus , Staphylococcus aureus
2.
Microbiome ; 9(1): 146, 2021 06 27.
Article in English | MEDLINE | ID: mdl-34176489

ABSTRACT

BACKGROUND: The maternal microbiome has emerged as an important factor in gestational health and outcome and is associated with risk of preterm birth and offspring morbidity. Epidemiological evidence also points to successive pregnancies-referred to as maternal parity-as a risk factor for preterm birth, infant mortality, and impaired neonatal growth. Despite the fact that both the maternal microbiome and parity are linked to maternal-infant health, the impact of parity on the microbiome remains largely unexplored, in part due to the challenges of studying parity in humans. RESULTS: Using synchronized pregnancies and dense longitudinal monitoring of the microbiome in pigs, we describe a microbiome trajectory during pregnancy and determine the extent to which parity modulates this trajectory. We show that the microbiome changes reproducibly during gestation and that this remodeling occurs more rapidly as parity increases. At the time of parturition, parity was linked to the relative abundance of several bacterial species, including Treponema bryantii, Lactobacillus amylovorus, and Lactobacillus reuteri. Strain tracking carried out in 18 maternal-offspring "quadrads"-each consisting of one mother sow and three piglets-linked maternal parity to altered levels of Akkermansia muciniphila, Prevotella stercorea, and Campylobacter coli in the infant gut 10 days after birth. CONCLUSIONS: Collectively, these results identify parity as an important environmental factor that modulates the gut microbiome during pregnancy and highlight the utility of a swine model for investigating the microbiome in maternal-infant health. In addition, our data show that the impact of parity extends beyond the mother and is associated with alterations in the community of bacteria that colonize the offspring gut early in life. The bacterial species we identified as parity-associated in the mother and offspring have been shown to influence host metabolism in other systems, raising the possibility that such changes may influence host nutrient acquisition or utilization. These findings, taken together with our observation that even subtle differences in parity are associated with microbiome changes, underscore the importance of considering parity in the design and analysis of human microbiome studies during pregnancy and in infants. Video abstract.


Subject(s)
Gastrointestinal Microbiome , Premature Birth , Animals , Female , Parity , Pregnancy , Prevotella , Swine , Treponema
3.
Sci Transl Med ; 11(519)2019 11 20.
Article in English | MEDLINE | ID: mdl-31748229

ABSTRACT

Patients infected with Leishmania braziliensis develop chronic lesions that often fail to respond to treatment with antiparasite drugs. To determine whether genes whose expression is highly variable in lesions between patients might influence disease outcome, we obtained biopsies of lesions from patients before treatment with pentavalent antimony and performed transcriptomic profiling on these clinical samples. We identified genes that were highly variably expressed between patients, and the variable expression of these genes correlated with treatment outcome. Among the most variable genes in all the patients were components of the cytolytic pathway, and the expression of these genes correlated with parasite load in the skin. We demonstrated that treatment failure was linked to the cytolytic pathway activated during infection. Using a host-pathogen marker profile of as few as three genes, we showed that eventual treatment outcome could be predicted before the start of treatment in two separate cohorts of patients with cutaneous leishmaniasis (n = 21 and n = 25). These findings raise the possibility of point-of-care diagnostic screening to identify patients at high risk of treatment failure and provide a rationale for a precision medicine approach to drug selection in cutaneous leishmaniasis. This work more broadly demonstrates the value of identifying genes of high variability in other diseases to better understand and predict diverse clinical outcomes.


Subject(s)
Gene Expression Regulation , Leishmaniasis, Cutaneous/drug therapy , Leishmaniasis, Cutaneous/genetics , Parasite Load , CD8-Positive T-Lymphocytes/immunology , Cell Death , Gene Expression Profiling , Humans , Killer Cells, Natural/immunology , Leishmania braziliensis/genetics , Leishmaniasis, Cutaneous/parasitology , Skin/parasitology , Skin/pathology , Treatment Outcome
4.
Microbiome ; 7(1): 126, 2019 08 31.
Article in English | MEDLINE | ID: mdl-31472697

ABSTRACT

BACKGROUND: The microbiome has been implicated in the initiation and persistence of inflammatory bowel disease. Despite the fact that diet is one of the most potent modulators of microbiome composition and function and that dietary intervention is the first-line therapy for treating pediatric Crohn's disease, the relationships between diet-induced remission, enteropathy, and microbiome are poorly understood. Here, we leverage a naturally-occurring canine model of chronic inflammatory enteropathy that exhibits robust remission following nutritional therapy, to perform a longitudinal study that integrates clinical monitoring, 16S rRNA gene amplicon sequencing, metagenomic sequencing, metabolomic profiling, and whole genome sequencing to investigate the relationship between therapeutic diet, microbiome, and disease. RESULTS: We show that remission induced by a hydrolyzed protein diet is accompanied by alterations in microbial community structure marked by decreased abundance of pathobionts (e.g., Escherichia coli and Clostridium perfringens), reduced severity of dysbiosis, and increased levels of the secondary bile acids, lithocholic and deoxycholic acid. Physiologic levels of these bile acids inhibited the growth of E. coli and C. perfringens isolates, in vitro. Metagenomic analysis and whole genome sequencing identified the bile acid producer Clostridium hiranonis as elevated after dietary therapy and a likely source of secondary bile acids during remission. When C. hiranonis was administered to mice, levels of deoxycholic acid were preserved and pathology associated with DSS colitis was ameliorated. Finally, a closely related bile acid producer, Clostridium scindens, was associated with diet-induced remission in human pediatric Crohn's disease. CONCLUSIONS: These data highlight that remission induced by a hydrolyzed protein diet is associated with improved microbiota structure, an expansion of bile acid-producing clostridia, and increased levels of secondary bile acids. Our observations from clinical studies of exclusive enteral nutrition in human Crohn's disease, along with our in vitro inhibition assays and in vivo studies in mice, suggest that this may be a conserved response to diet therapy with the potential to ameliorate disease. These findings provide insight into diet-induced remission of gastrointestinal disease and could help guide the rational design of more effective therapeutic diets.


Subject(s)
Bile Acids and Salts/metabolism , Crohn Disease/microbiology , Diet Therapy/methods , Dysbiosis , Gastrointestinal Microbiome , Animals , Child , Clostridiales/metabolism , Dogs , Dysbiosis/microbiology , Dysbiosis/therapy , Humans , Longitudinal Studies , Male , Mice , Mice, Inbred C57BL , Remission Induction
5.
mBio ; 10(3)2019 05 28.
Article in English | MEDLINE | ID: mdl-31138751

ABSTRACT

Oral infection of C57BL/6J mice with Toxoplasma gondii results in a marked bacterial dysbiosis and the development of severe pathology in the distal small intestine that is dependent on CD4+ T cells and interferon gamma (IFN-γ). This dysbiosis and bacterial translocation contribute to the development of ileal pathology, but the factors that support the bloom of bacterial pathobionts are unclear. The use of microbial community profiling and shotgun metagenomics revealed that Toxoplasma infection induces a dysbiosis dominated by Enterobacteriaceae and an increased potential for nitrate respiration. In vivo experiments using bacterial metabolic mutants revealed that during this infection, host-derived nitrate supports the expansion of Enterobacteriaceae in the ileum via nitrate respiration. Additional experiments with infected mice indicate that the IFN-γ/STAT1/iNOS axis, while essential for parasite control, also supplies a pool of nitrate that serves as a source for anaerobic respiration and supports overgrowth of Enterobacteriaceae Together, these data reveal a trade-off in intestinal immunity after oral infection of C57BL/6J mice with T. gondii, in which inducible nitric oxide synthase (iNOS) is required for parasite control, while this host enzyme is responsible for specific modification of the composition of the microbiome that contributes to pathology.IMPORTANCEToxoplasma gondii is a protozoan parasite and a leading cause of foodborne illness. Infection is initiated when the parasite invades the intestinal epithelium, and in many host species, this leads to intense inflammation and a dramatic disruption of the normal microbial ecosystem that resides in the healthy gut (the so-called microbiome). One characteristic change in the microbiome during infection with Toxoplasma-as well as numerous other pathogens-is the overgrowth of Escherichia coli or similar bacteria and a breakdown of commensal containment leading to seeding of peripheral organs with gut bacteria and subsequent sepsis. Our findings provide one clear explanation for how this process is regulated, thereby improving our understanding of the relationship between parasite infection, inflammation, and disease. Furthermore, our results could serve as the basis for the development of novel therapeutics to reduce the potential for harmful bacteria to bloom in the gut during infection.


Subject(s)
Dysbiosis/immunology , Gastrointestinal Microbiome , Intestine, Small/immunology , Intestine, Small/pathology , Macrophage Activation , Nitrates/metabolism , Toxoplasmosis, Animal/immunology , Animals , Cytokines/analysis , Enterobacteriaceae/classification , Female , Inflammation , Interferon-gamma/immunology , Intestine, Small/microbiology , Intestine, Small/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/immunology , Toxoplasma/pathogenicity , Toxoplasmosis, Animal/microbiology
6.
Nat Neurosci ; 21(8): 1061-1071, 2018 08.
Article in English | MEDLINE | ID: mdl-29988069

ABSTRACT

Early prenatal stress disrupts maternal-to-offspring microbiota transmission and has lasting effects on metabolism, physiology, cognition, and behavior in male mice. Here we show that transplantation of maternal vaginal microbiota from stressed dams into naive pups delivered by cesarean section had effects that partly resembled those seen in prenatally stressed males. However, transplantation of control maternal vaginal microbiota into prenatally stressed pups delivered by cesarean section did not rescue the prenatal-stress phenotype. Prenatal stress was associated with alterations in the fetal intestinal transcriptome and niche, as well as with changes in the adult gut that were altered by additional stress exposure in adulthood. Further, maternal vaginal transfer also partially mediated the effects of prenatal stress on hypothalamic gene expression, as observed after chronic stress in adulthood. These findings suggest that the maternal vaginal microbiota contribute to the lasting effects of prenatal stress on gut and hypothalamus in male mice.


Subject(s)
Gastrointestinal Tract/physiology , Hypothalamus/physiology , Microbiota , Prenatal Exposure Delayed Effects , Stress, Psychological/physiopathology , Vagina/microbiology , Animals , Body Weight , Cesarean Section , Female , Gastrointestinal Tract/embryology , Gene Expression/genetics , Hypothalamo-Hypophyseal System , Hypothalamus/metabolism , Intestinal Absorption , Male , Mice , Mice, Inbred C57BL , Paraventricular Hypothalamic Nucleus/metabolism , Phenotype , Pregnancy , Transcriptome
7.
Comp Med ; 68(4): 261-268, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29898804

ABSTRACT

Immunocompromised mice are used frequently in biomedical research, in part because they accommodate the engraftment and study of primary human cells within a mouse model; however, these animals are susceptible to opportunistic infections and require special husbandry considerations. In 2015, an outbreak marked by high morbidity but low mortality swept through a colony of immunocompromised mice; this outbreak rapidly affected 75% of the colony and ultimately required complete depopulation of the barrier suite. Conventional microbiologic and molecular diagnostics were unsuccessful in determining the cause; therefore, we explored culture-independent methods to broadly profile the microbial community in the feces of affected animals. This approach identified 4 bacterial taxa- Candidatus Arthromitus, Clostridium celatum, Clostridiales bacterium VE202-01, and Bifidobacterium pseudolongum strain PV8-2- that were significantly enriched in the affected mice. Based on these results, specific changes were made to the animal husbandry procedures for immunocompromised mice. This case report highlights the utility of culture-independent methods in laboratory animal diagnostics.


Subject(s)
Diarrhea/microbiology , Feces/microbiology , Microbiota/genetics , Animals , Diarrhea/epidemiology , Disease Outbreaks , Genetic Variation , Immunocompromised Host , Metagenomics , Mice , Mice, Inbred NOD , Mice, SCID , RNA, Ribosomal, 16S/chemistry , Sequence Analysis, RNA , Whole Genome Sequencing
8.
Cell Host Microbe ; 23(3): 302-311.e3, 2018 Mar 14.
Article in English | MEDLINE | ID: mdl-29478774

ABSTRACT

Serum immunoglobulin A (IgA) antibodies are readily detected in mice and people, but the mechanisms underlying the induction of serum IgA and its role in host protection remain uncertain. We report that select commensal bacteria induce several facets of systemic IgA-mediated immunity. Exposing conventional mice to a unique but natural microflora that included several members of the Proteobacteria phylum led to T cell-dependent increases in serum IgA levels and the induction of large numbers of IgA-secreting plasma cells in the bone marrow. The resulting serum IgA bound to a restricted collection of bacterial taxa, and antigen-specific serum IgA antibodies were readily induced after intestinal colonization with the commensal bacterium Helicobacter muridarum. Finally, movement to a Proteobacteria-rich microbiota led to serum IgA-mediated resistance to polymicrobial sepsis. We conclude that commensal microbes overtly influence the serum IgA repertoire, resulting in constitutive protection against bacterial sepsis.


Subject(s)
Gastrointestinal Microbiome/immunology , Immunoglobulin A/blood , Immunoglobulin A/immunology , Microbiota/immunology , Sepsis/immunology , Sepsis/microbiology , Symbiosis , Animals , Antigens, Bacterial/blood , Antigens, Bacterial/immunology , B-Lymphocytes/immunology , Bacteria/classification , Bacteria/genetics , Bacteria/immunology , Bone Marrow , Female , Intestinal Mucosa/microbiology , Intestines/immunology , Intestines/microbiology , Mice , Mice, Inbred C57BL , Plasma Cells/immunology , Proteobacteria/classification , Proteobacteria/immunology , Proteobacteria/physiology , RNA, Ribosomal, 16S/genetics , T-Lymphocytes/immunology
9.
Cell Host Microbe ; 22(1): 13-24.e4, 2017 Jul 12.
Article in English | MEDLINE | ID: mdl-28669672

ABSTRACT

Skin microbiota can impact allergic and autoimmune responses, wound healing, and anti-microbial defense. We investigated the role of skin microbiota in cutaneous leishmaniasis and found that human patients infected with Leishmania braziliensis develop dysbiotic skin microbiota, characterized by increases in the abundance of Staphylococcus and/or Streptococcus. Mice infected with L. major exhibit similar changes depending upon disease severity. Importantly, this dysbiosis is not limited to the lesion site, but is transmissible to normal skin distant from the infection site and to skin from co-housed naive mice. This observation allowed us to test whether a pre-existing dysbiotic skin microbiota influences disease, and we found that challenging dysbiotic naive mice with L. major or testing for contact hypersensitivity results in exacerbated skin inflammatory responses. These findings demonstrate that a dysbiotic skin microbiota is not only a consequence of tissue stress, but also enhances inflammation, which has implications for many inflammatory cutaneous diseases.


Subject(s)
Dysbiosis/etiology , Dysbiosis/immunology , Inflammation , Leishmania braziliensis/pathogenicity , Leishmaniasis, Cutaneous/complications , Leishmaniasis, Cutaneous/microbiology , Microbiota/physiology , Skin/immunology , Animals , Disease Models, Animal , Humans , Hypersensitivity , Inflammation/immunology , Inflammation/microbiology , Leishmania major/immunology , Leishmania major/pathogenicity , Mice , Mice, Inbred C57BL , Microbiota/immunology , Skin/microbiology , Skin/parasitology , Staphylococcus/immunology , Staphylococcus/pathogenicity , Streptococcus/immunology , Streptococcus/pathogenicity
10.
Sci Rep ; 7: 44182, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28266645

ABSTRACT

The microbiome is a regulator of host immunity, metabolism, neurodevelopment, and behavior. During early life, bacterial communities within maternal gut and vaginal compartments can have an impact on directing these processes. Maternal stress experience during pregnancy may impact offspring development by altering the temporal and spatial dynamics of the maternal microbiome during pregnancy. To examine the hypothesis that maternal stress disrupts gut and vaginal microbial dynamics during critical prenatal and postnatal windows, we used high-resolution 16S rRNA marker gene sequencing to examine outcomes in our mouse model of early prenatal stress. Consistent with predictions, maternal fecal communities shift across pregnancy, a process that is disrupted by stress. Vaginal bacterial community structure and composition exhibit lasting disruption following stress exposure. Comparison of maternal and offspring microbiota revealed that similarities in bacterial community composition was predicted by a complex interaction between maternal body niche and offspring age and sex. Importantly, early prenatal stress influenced offspring bacterial community assembly in a temporal and sex-specific manner. Taken together, our results demonstrate that early prenatal stress may influence offspring development through converging modifications to gut microbial composition during pregnancy and transmission of dysbiotic vaginal microbiome at birth.


Subject(s)
Microbiota , Pregnancy Complications/microbiology , Sex Characteristics , Stress, Psychological/microbiology , Animals , Female , Male , Mice , Pregnancy
11.
mSphere ; 1(5)2016.
Article in English | MEDLINE | ID: mdl-27704053

ABSTRACT

Staphylococcus species are a leading cause of skin and soft tissue infections in humans and animals, and the antibiotics used to treat these infections are often the same. Methicillin- and multidrug-resistant staphylococcal infections are becoming more common in human and veterinary medicine. From a "One Health" perspective, this overlap in antibiotic use and resistance raises concerns over the potential spread of antibiotic resistance genes. Whole-genome sequencing and comparative genomics analysis revealed that Staphylococcus species use divergent pathways to synthesize isoprenoids. Species frequently associated with skin and soft tissue infections in companion animals, including S. schleiferi and S. pseudintermedius, use the nonmevalonate pathway. In contrast, S. aureus, S. epidermidis, and S. lugdunensis use the mevalonate pathway. The antibiotic fosmidomycin, an inhibitor of the nonmevalonate pathway, was effective in killing canine clinical staphylococcal isolates but had no effect on the growth or survival of S. aureus and S. epidermidis. These data identify an essential metabolic pathway in Staphylococcus that differs among members of this genus and suggest that drugs such as fosmidomycin, which targets enzymes in the nonmevalonate pathway, may be an effective treatment for certain staphylococcal infections. IMPORTANCE Drug-resistant Staphylococcus species are a major concern in human and veterinary medicine. There is a need for new antibiotics that exhibit a selective effect in treating infections in companion and livestock animals and that would not be used to treat human bacterial infections. We have identified fosmidomycin as an antibiotic that selectively targets certain Staphylococcus species that are often encountered in skin infections in cats and dogs. These findings expand our understanding of Staphylococcus evolution and may have direct implications for treating staphylococcal infections in veterinary medicine.

12.
Nature ; 535(7612): 440-443, 2016 07 21.
Article in English | MEDLINE | ID: mdl-27409807

ABSTRACT

Group 3 innate lymphoid cells (ILC3) are major regulators of inflammation and infection at mucosal barriers. ILC3 development is thought to be programmed, but how ILC3 perceive, integrate and respond to local environmental signals remains unclear. Here we show that ILC3 in mice sense their environment and control gut defence as part of a glial­ILC3­epithelial cell unit orchestrated by neurotrophic factors. We found that enteric ILC3 express the neuroregulatory receptor RET. ILC3-autonomous Ret ablation led to decreased innate interleukin-22 (IL-22), impaired epithelial reactivity, dysbiosis and increased susceptibility to bowel inflammation and infection. Neurotrophic factors directly controlled innate Il22 downstream of the p38 MAPK/ERK-AKT cascade and STAT3 activation. Notably, ILC3 were adjacent to neurotrophic-factor-expressing glial cells that exhibited stellate-shaped projections into ILC3 aggregates. Glial cells sensed microenvironmental cues in a MYD88-dependent manner to control neurotrophic factors and innate IL-22. Accordingly, glial-intrinsic Myd88 deletion led to impaired production of ILC3-derived IL-22 and a pronounced propensity towards gut inflammation and infection. Our work sheds light on a novel multi-tissue defence unit, revealing that glial cells are central hubs of neuron and innate immune regulation by neurotrophic factor signals.


Subject(s)
Immunity, Innate , Intestines/immunology , Lymphocytes/immunology , Neuroglia/metabolism , Neurotransmitter Agents/metabolism , Animals , Cellular Microenvironment/immunology , Epithelial Cells/cytology , Epithelial Cells/immunology , Female , Gastrointestinal Microbiome/immunology , Immunity, Mucosal , Inflammation/immunology , Inflammation/metabolism , Interleukins/biosynthesis , Interleukins/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestines/cytology , Lymphocytes/cytology , Lymphocytes/metabolism , MAP Kinase Signaling System , Male , Mice , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/metabolism , Neuroglia/immunology , Neurotransmitter Agents/immunology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-ret/deficiency , Proto-Oncogene Proteins c-ret/metabolism , STAT3 Transcription Factor/metabolism , Interleukin-22
13.
J Invest Dermatol ; 136(6): 1182-1190, 2016 06.
Article in English | MEDLINE | ID: mdl-26854488

ABSTRACT

Host-microbe interactions may play a fundamental role in the pathogenesis of atopic dermatitis, a chronic relapsing inflammatory skin disorder characterized by universal colonization with Staphylococcus species. To examine the relationship between epidermal barrier function and the cutaneous microbiota in atopic dermatitis, this study used a spontaneous model of canine atopic dermatitis. In a cohort of 14 dogs with canine atopic dermatitis, the skin microbiota were longitudinally evaluated with parallel assessment of skin barrier function at disease flare, during antimicrobial therapy, and post-therapy. Sequencing of the bacterial 16S ribosomal RNA gene showed decreased bacterial diversity and increased proportions of Staphylococcus (S. pseudintermedius in particular) and Corynebacterium species compared with a cohort of healthy control dogs (n = 16). Treatment restored bacterial diversity with decreased proportions of Staphylococcus species, concurrent with decreased canine atopic dermatitis severity. Skin barrier function, as measured by corneometry, pH, and transepidermal water loss also normalized with treatment. Bacterial diversity correlated with transepidermal water loss and pH level but not with corneometry results. These findings provide insights into the relationship between the cutaneous microbiome and skin barrier function in atopic dermatitis, show the impact of antimicrobial therapy on the skin microbiome, and highlight the utility of canine atopic dermatitis as a spontaneous nonrodent model of atopic dermatitis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/microbiology , Microbiota/drug effects , Staphylococcus/pathogenicity , Animals , Biopsy, Needle , Dermatitis, Atopic/pathology , Disease Models, Animal , Dog Diseases/drug therapy , Dog Diseases/pathology , Dogs , Female , Immunohistochemistry , Longitudinal Studies , Male , Random Allocation , Statistics, Nonparametric , Treatment Outcome
14.
Genome Announc ; 3(5)2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26358596

ABSTRACT

Staphylococcus schleiferi, a Gram-positive and coagulase-variable organism, is an opportunistic human pathogen and a major cause of skin and soft tissue infections in dogs. Here, we report the first S. schleiferi genome sequence and methylome from four canine clinical isolates.

15.
Vet Microbiol ; 180(3-4): 304-7, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26411322

ABSTRACT

The strain dynamics of methicillin-resistant Staphylococcus aureus (MRSA) isolates from people and the household dog were investigated. The isolates were identified in the context of a randomized controlled trial that tested household-wide decolonization of people. Genotypic comparison of MRSA isolates obtained from two household members, the dog, and home surfaces over a three-month period failed to implicate the pet or the home environment in recurrent colonization of the household members. However, it did implicate the pet's bed in exposure of the dog prior to the dog's infection. Whole genome sequencing was performed to differentiate the isolates. This report also describes introduction of diverse strains of MRSA into the household within six weeks of cessation of harmonized decolonization treatment of people and treatment for infection in the dog. These findings suggest that community sources outside the home may be important for recurrent MRSA colonization or infection.


Subject(s)
Dogs/microbiology , Genome, Bacterial , Methicillin-Resistant Staphylococcus aureus/genetics , Staphylococcal Infections/veterinary , Adult , Animals , Bacterial Typing Techniques , Bedding and Linens , Female , Genotype , Humans , Male , Methicillin-Resistant Staphylococcus aureus/classification , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Randomized Controlled Trials as Topic
16.
Microbiome ; 3: 2, 2015.
Article in English | MEDLINE | ID: mdl-25705378

ABSTRACT

BACKGROUND: Staphylococcus aureus and other coagulase-positive staphylococci (CPS) colonize skin and mucous membrane sites and can cause skin and soft tissue infections (SSTIs) in humans and animals. Factors modulating methicillin-resistant S. aureus (MRSA) colonization and infection in humans remain unclear, including the role of the greater microbial community and environmental factors such as contact with companion animals. In the context of a parent study evaluating the households of outpatients with community MRSA SSTI, the objectives of this study were 1) to characterize the microbiota that colonizes typical coagulase-positive Staphylococcus spp. carriage sites in humans and their companion pets, 2) to analyze associations between Staphylococcus infection and carriage and the composition and diversity of microbial communities, and 3) to analyze factors that influence sharing of microbiota between pets and humans. RESULTS: We enrolled 25 households containing 56 pets and 30 humans. Sampling locations were matched to anatomical sites cultured by the parent study for MRSA and other CPS. Bacterial microbiota were characterized by sequencing of 16S ribosomal RNA genes. Household membership was strongly associated with microbial communities, in both humans and pets. Pets were colonized with a greater relative abundance of Proteobacteria, whereas people were colonized with greater relative abundances of Firmicutes and Actinobacteria. We did not detect differences in microbiota associated with MRSA SSTI, or carriage of MRSA, S. aureus or CPS. Humans in households without pets were more similar to each other than humans in pet-owning households, suggesting that companion animals may play a role in microbial transfer. We examined changes in microbiota over a 3-month time period and found that pet staphylococcal carriage sites were more stable than human carriage sites. CONCLUSIONS: We characterized and identified patterns of microbiota sharing and stability between humans and companion animals. While we did not detect associations with MRSA SSTI, or carriage of MRSA, S. aureus or CPS in this small sample size, larger studies are warranted to fully explore how microbial communities may be associated with and contribute to MRSA and/or CPS colonization, infection, and recurrence.

17.
Adv Wound Care (New Rochelle) ; 3(7): 502-510, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-25032070

ABSTRACT

Significance: Bacterial burden is believed to play a significant role in impaired wound healing of chronic wounds and the development of infection-related complications. The standard of care in the clinic relies upon cultivation-dependent methods to identify microorganisms. These assays are biased toward microorganisms that thrive in isolation under laboratory conditions. Recent Advances: Significant advances in genomic technologies have enabled less-biased, culture-independent approaches to characterize microbial communities, or microbiomes. The aggregate sequencing and analysis of 16S ribosomal RNA genes has demonstrated that cultures under-represent true microbial diversity and load. Critical Issues: Despite recent advances that enable culture-independent analyses of microbiomes, those organisms that are important in impaired healing remain ambiguous. Inconsistent findings across various studies highlight the need to characterize microbiomes of chronic wounds with homogenous etiology to determine differences in microbiomes that may be driven by the wound environment and that may affect wound outcomes. Rigorous analyses of wound microbiomes in light of the three dimensions of bioburden (microbial diversity, microbial load, and pathogenic organisms), clinical metadata, and wound outcomes will be a significant step forward in our quest to understand the role of microorganisms in impaired healing. Future Directions: Longitudinal studies employing serial sampling are needed to appreciate the role of the dynamic microbial community in chronic wound healing. The value of clinical metadata needs to be examined as potential biomarkers of problematic microbiota and wound outcomes. Lastly, better characterization and understanding of wound microbiomes will open avenues for improved diagnostic and therapeutic tools for the nonhealing wound.

18.
J Mol Biol ; 400(5): 1011-21, 2010 Jul 30.
Article in English | MEDLINE | ID: mdl-20595000

ABSTRACT

Type IV pili are bacterial extracellular filaments that can be retracted to create force and motility. Retraction is accomplished by the motor protein PilT. Crystal structures of Pseudomonas aeruginosa PilT with and without bound beta,gamma-methyleneadenosine-5'-triphosphate have been solved at 2.6 A and 3.1 A resolution, respectively, revealing an interlocking hexamer formed by the action of a crystallographic 2-fold symmetry operator on three subunits in the asymmetric unit and held together by extensive ionic interactions. The roles of two invariant carboxylates, Asp Box motif Glu163 and Walker B motif Glu204, have been assigned to Mg(2+) binding and catalysis, respectively. The nucleotide ligands in each of the subunits in the asymmetric unit of the beta,gamma-methyleneadenosine-5'-triphosphate-bound PilT are not equally well ordered. Similarly, the three subunits in the asymmetric unit of both structures exhibit differing relative conformations of the two domains. The 12 degrees and 20 degrees domain rotations indicate motions that occur during the ATP-coupled mechanism of the disassembly of pili into membrane-localized pilin monomers. Integrating these observations, we propose a three-state "Ready, Active, Release" model for the action of PilT.


Subject(s)
Fimbriae, Bacterial/chemistry , Nucleotides/chemistry , Pseudomonas aeruginosa/chemistry , Binding Sites , Crystallography, X-Ray , Ligands , Protein Conformation
19.
Structure ; 15(3): 363-76, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17355871

ABSTRACT

PilT is a hexameric ATPase required for bacterial type IV pilus retraction and surface motility. Crystal structures of ADP- and ATP-bound Aquifex aeolicus PilT at 2.8 and 3.2 A resolution show N-terminal PAS-like and C-terminal RecA-like ATPase domains followed by a set of short C-terminal helices. The hexamer is formed by extensive polar subunit interactions between the ATPase core of one monomer and the N-terminal domain of the next. An additional structure captures a nonsymmetric PilT hexamer in which approach of invariant arginines from two subunits to the bound nucleotide forms an enzymatically competent active site. A panel of pilT mutations highlights the importance of the arginines, the PAS-like domain, the polar subunit interface, and the C-terminal helices for retraction. We present a model for ATP binding leading to dramatic PilT domain motions, engagement of the arginine wire, and subunit communication in this hexameric motor. Our conclusions apply to the entire type II/IV secretion ATPase family.


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/physiology , Bacterial Proteins/chemistry , Bacterial Proteins/physiology , Fimbriae, Bacterial/physiology , Molecular Motor Proteins/chemistry , Molecular Motor Proteins/physiology , Movement/physiology , Protein Subunits/chemistry , Protein Subunits/physiology , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Arginine/chemistry , Bacterial Proteins/genetics , Crystallography, X-Ray , Fimbriae, Bacterial/genetics , Molecular Motor Proteins/genetics , Molecular Sequence Data , Protein Structure, Tertiary/genetics , Protein Subunits/genetics
20.
Proc Natl Acad Sci U S A ; 101(23): 8575-80, 2004 Jun 08.
Article in English | MEDLINE | ID: mdl-15159541

ABSTRACT

Single-stranded DNA (ssDNA)-binding (SSB) proteins are uniformly required to bind and protect single-stranded intermediates in DNA metabolic pathways. All bacterial and eukaryotic SSB proteins studied to date oligomerize to assemble four copies of a conserved domain, called an oligonucleotide/oligosaccharide-binding (OB) fold, that cooperate in nonspecific ssDNA binding. The vast majority of bacterial SSB family members function as homotetramers, with each monomer contributing a single OB fold. However, SSB proteins from the Deinococcus-Thermus genera are exceptions to this rule, because they contain two OB folds per monomer. To investigate the structural consequences of this unusual arrangement, we have determined a 1.8-A-resolution x-ray structure of Deinococcus radiodurans SSB. The structure shows that D. radiodurans SSB comprises two OB domains linked by a beta-hairpin motif. The protein assembles a four-OB-fold arrangement by means of symmetric dimerization. In contrast to homotetrameric SSB proteins, asymmetry exists between the two OB folds of D. radiodurans SSB because of sequence differences between the domains. These differences appear to reflect specialized roles that have evolved for each domain. Extensive crystallographic contacts link D. radiodurans SSB dimers in an arrangement that has important implications for higher-order structures of the protein bound to ssDNA. This assembly utilizes the N-terminal OB domain and the beta-hairpin structure that is unique to Deinococcus and Thermus species SSB proteins. We hypothesize that differences between D. radiodurans SSB and homotetrameric bacterial SSB proteins may confer a selective advantage to D. radiodurans cells that aids viability in environments that challenge genomic stability.


Subject(s)
Bacterial Proteins/chemistry , DNA-Binding Proteins/chemistry , Deinococcus/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Crystallography, X-Ray , DNA Damage , DNA, Bacterial/metabolism , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Deinococcus/genetics , Deinococcus/metabolism , Dimerization , Models, Molecular , Molecular Sequence Data , Protein Structure, Quaternary , Protein Structure, Tertiary , Static Electricity
SELECTION OF CITATIONS
SEARCH DETAIL
...