Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Cell Host Microbe ; 29(8): 1235-1248.e8, 2021 Aug 11.
Article in English | MEDLINE | ID: mdl-34214492

ABSTRACT

The epidermis forms a barrier that defends the body from desiccation and entry of harmful substances, while also sensing and integrating environmental signals. The tightly orchestrated cellular changes needed for the formation and maintenance of this epidermal barrier occur in the context of the skin microbiome. Using germ-free mice, we demonstrate the microbiota is necessary for proper differentiation and repair of the epidermal barrier. These effects are mediated by microbiota signaling through the aryl hydrocarbon receptor (AHR) in keratinocytes, a xenobiotic receptor also implicated in epidermal differentiation. Mice lacking keratinocyte AHR are more susceptible to barrier damage and infection, during steady-state and epicutaneous sensitization. Colonization with a defined consortium of human skin isolates restored barrier competence in an AHR-dependent manner. We reveal a fundamental mechanism whereby the microbiota regulates skin barrier formation and repair, which has far-reaching implications for the numerous skin disorders characterized by epidermal barrier dysfunction.


Subject(s)
Microbiota/physiology , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Skin/microbiology , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation , Cell Line , Epidermal Cells/metabolism , Epidermal Cells/pathology , Epidermis/metabolism , Female , Humans , Keratinocytes , Male , Mice , Mice, Inbred C57BL , Skin/pathology , Skin Diseases/microbiology
2.
Vet Dermatol ; 31(3): 225-e49, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31960536

ABSTRACT

BACKGROUND: Canine otitis externa (OE) is a common inflammatory disease that is frequently complicated by secondary bacterial and/or yeast infections. The otic microbial population is more complex than appreciated by cytological methods and aerobic culture alone. HYPOTHESIS/OBJECTIVES: Differences in bacterial and fungal populations of the external ear canal will correlate with specific cytological and culture-based definitions of bacterial and Malassezia otitis. ANIMALS: Forty client-owned dogs; 30 with OE and 10 with healthy ears. METHODS AND MATERIALS: Prospective study comparing cytological samples, aerobic bacterial cultures and culture-independent sequencing-based analyses of the external ear canal. Subjects with OE included 10 dogs with only cocci [≥25/high power field (HPF)] on cytological evaluation and culture of Staphylococcus spp.; 10 dogs with rods (≥25/HPF) and exclusive culture of Pseudomonas aeruginosa; 10 dogs with only yeast on cytological results morphologically compatible with Malassezia spp. (≥5/HPF). RESULTS: Staphylococcus was the most abundant taxa across all groups. Ears cytologically positive for cocci had decreased diversity, and all types of OE were associated with decreased fungal diversity compared to controls. CONCLUSIONS AND CLINICAL IMPORTANCE: Cytological and culture-based assessment of the ear canal is not predictive of the diverse microbiota of the ear canal in cases of Pseudomonas or Malassezia otitis. Less abundant bacterial taxa in cases of staphylococcal OE are worth scrutiny for future biological therapy.


Subject(s)
Dog Diseases/microbiology , Ear Canal/microbiology , Microbiota , Mycobiome , Otitis Externa/microbiology , Animals , Bacteria/classification , Bacteria/isolation & purification , Dog Diseases/epidemiology , Dogs , Ear Canal/pathology , Female , Fungi/classification , Fungi/isolation & purification , Malassezia/pathogenicity , Male , Otitis Externa/epidemiology , Prospective Studies , Pseudomonas/pathogenicity , United States/epidemiology
3.
Cell Host Microbe ; 25(5): 641-655.e5, 2019 May 08.
Article in English | MEDLINE | ID: mdl-31006638

ABSTRACT

Chronic wounds are a major complication of diabetes associated with high morbidity and health care expenditures. To investigate the role of colonizing microbiota in diabetic wound healing, clinical outcomes, and response to interventions, we conducted a longitudinal, prospective study of patients with neuropathic diabetic foot ulcers (DFU). Metagenomic shotgun sequencing revealed that strain-level variation of Staphylococcus aureus and genetic signatures of biofilm formation were associated with poor outcomes. Cultured wound isolates of S. aureus elicited differential phenotypes in mouse models that corresponded with patient outcomes, while wound "bystanders" such as Corynebacterium striatum and Alcaligenes faecalis, typically considered commensals or contaminants, also significantly impacted wound severity and healing. Antibiotic resistance genes were widespread, and debridement, rather than antibiotic treatment, significantly shifted the DFU microbiota in patients with more favorable outcomes. These findings suggest that the DFU microbiota may be a marker for clinical outcomes and response to therapeutic interventions.


Subject(s)
Anti-Infective Agents/therapeutic use , Coinfection/microbiology , Debridement , Diabetic Foot/microbiology , Microbiota , Wound Infection/microbiology , Animals , Coinfection/therapy , Diabetic Foot/therapy , Disease Models, Animal , Longitudinal Studies , Mice , Prospective Studies , Treatment Outcome , Wound Healing , Wound Infection/therapy
4.
BMC Microbiol ; 18(1): 174, 2018 11 03.
Article in English | MEDLINE | ID: mdl-30390618

ABSTRACT

BACKGROUND: We sought to determine if the prevalence of antibiotic-resistant Escherichia coli differed across retail poultry products and among major production categories, including organic, "raised without antibiotics", and conventional. RESULTS: We collected all available brands of retail chicken and turkey-including conventional, "raised without antibiotic", and organic products-every two weeks from January to December 2012. In total, E. coli was recovered from 91% of 546 turkey products tested and 88% of 1367 chicken products tested. The proportion of samples contaminated with E. coli was similar across all three production categories. Resistance prevalence varied by meat type and was highest among E. coli isolates from turkey for the majority of antibiotics tested. In general, production category had little effect on resistance prevalence among E. coli isolates from chicken, although resistance to gentamicin and multidrug resistance did vary. In contrast, resistance prevalence was significantly higher for 6 of the antibiotics tested-and multidrug resistance-among isolates from conventional turkey products when compared to those labelled organic or "raised without antibiotics". E. coli isolates from chicken varied strongly in resistance prevalence among different brands within each production category. CONCLUSION: The high prevalence of resistance among E. coli isolates from conventionally-raised turkey meat suggests greater antimicrobial use in conventional turkey production as compared to "raised without antibiotics" and organic systems. However, among E. coli from chicken meat, resistance prevalence was more strongly linked to brand than to production category, which could be caused by brand-level differences during production and/or processing, including variations in antimicrobial use.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial , Escherichia coli/drug effects , Food Microbiology , Food, Organic/microbiology , Poultry/microbiology , Animals , Chickens/microbiology , Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli/genetics , Food Contamination , Genes, Bacterial/genetics , Microbial Sensitivity Tests , Turkeys/microbiology
5.
mBio ; 9(4)2018 08 28.
Article in English | MEDLINE | ID: mdl-30154256

ABSTRACT

Escherichia coli sequence type 131 (ST131) has emerged rapidly to become the most prevalent extraintestinal pathogenic E. coli clones in circulation today. Previous investigations appeared to exonerate retail meat as a source of human exposure to ST131; however, these studies focused mainly on extensively multidrug-resistant ST131 strains, which typically carry allele 30 of the fimH type 1 fimbrial adhesin gene (ST131-H30). To estimate the frequency of extraintestinal human infections arising from foodborne ST131 strains without bias toward particular sublineages or phenotypes, we conducted a 1-year prospective study of E. coli from meat products and clinical cultures in Flagstaff, Arizona. We characterized all isolates by multilocus sequence typing, fimH typing, and core genome phylogenetic analyses, and we screened isolates for avian-associated ColV plasmids as an indication of poultry adaptation. E. coli was isolated from 79.8% of the 2,452 meat samples and 72.4% of the 1,735 culture-positive clinical samples. Twenty-seven meat isolates were ST131 and belonged almost exclusively (n = 25) to the ST131-H22 lineage. All but 1 of the 25 H22 meat isolates were from poultry products, and all but 2 carried poultry-associated ColV plasmids. Of the 1,188 contemporaneous human clinical E. coli isolates, 24 were ST131-H22, one-quarter of which occurred in the same high-resolution phylogenetic clades as the ST131-H22 meat isolates and carried ColV plasmids. Molecular clock analysis of an international ST131-H22 genome collection suggested that ColV plasmids have been acquired at least six times since the 1940s and that poultry-to-human transmission is not limited to the United States.IMPORTANCEE. coli ST131 is an important extraintestinal pathogen that can colonize the gastrointestinal tracts of humans and food animals. Here, we combined detection of accessory traits associated with avian adaptation (ColV plasmids) with high-resolution phylogenetics to quantify the portion of human infections caused by ST131 strains of food animal origin. Our results suggest that one ST131 sublineage-ST131-H22-has become established in poultry populations around the world and that meat may serve as a vehicle for human exposure and infection. ST131-H22 is just one of many E. coli lineages that may be transmitted from food animals to humans. Additional studies that combine detection of host-associated accessory elements with phylogenetics may allow us to quantify the total fraction of human extraintestinal infections attributable to food animal E. coli strains.


Subject(s)
Escherichia coli Infections/microbiology , Meat/microbiology , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/isolation & purification , Adhesins, Escherichia coli/genetics , Arizona , Fimbriae Proteins/genetics , Humans , Multilocus Sequence Typing , Phylogeny , Plasmids/analysis , Prospective Studies , Uropathogenic Escherichia coli/classification , Uropathogenic Escherichia coli/genetics
6.
J Invest Dermatol ; 138(10): 2234-2243, 2018 10.
Article in English | MEDLINE | ID: mdl-29753031

ABSTRACT

Despite critical functions in cutaneous health and disease, it is unclear how resident skin microbial communities are altered by topical antimicrobial interventions commonly used in personal and clinical settings. Here we show that acute exposure to antiseptic treatments elicits rapid but short-term depletion of microbial community diversity and membership. Thirteen subjects were enrolled in a longitudinal treatment study to analyze the effects of topical treatments (i.e., ethanol, povidone-iodine, chlorhexidine, and water) on the skin microbiome at two skin sites of disparate microenvironment: forearm and back. Treatment effects were highly dependent on personalized and body site-specific colonization signatures, which concealed community dynamics at the population level when not accounted for in this analysis. The magnitude of disruption was influenced by the identity and abundance of particular bacterial inhabitants. Lowly abundant members of the skin microbiota were more likely to be displaced, and subsequently replaced, by the most abundant taxa prior to treatment. Members of the skin commensal family Propionibactericeae were particularly resilient to treatment, suggesting a distinct competitive advantage in the face of disturbance. These results provide insight into the stability and resilience of the skin microbiome, while establishing the impact of topical antiseptic treatment on skin bacterial dynamics and community ecology.


Subject(s)
Anti-Infective Agents, Local/administration & dosage , Bacteria/genetics , DNA, Bacterial/analysis , Microbiota/drug effects , Skin Diseases, Bacterial/prevention & control , Skin/microbiology , Administration, Cutaneous , Adult , Bacteria/isolation & purification , Female , Healthy Volunteers , Humans , Male , Polymerase Chain Reaction , Skin/drug effects , Skin Diseases, Bacterial/microbiology , Young Adult
7.
Wound Repair Regen ; 26(2): 127-135, 2018 03.
Article in English | MEDLINE | ID: mdl-29802752

ABSTRACT

Open fractures are characterized by disruption of the skin and soft tissue, which allows for microbial contamination and colonization. Preventing infection-related complications of open fractures and other acute wounds remains an evolving challenge due to an incomplete understanding of how microbial colonization and contamination influence healing and outcomes. Culture-independent molecular methods are now widely used to study human-associated microbial communities without introducing culture biases. Using such approaches, the objectives of this study were to (1) define the long-term temporal microbial community dynamics of open fracture wounds and (2) examine microbial community dynamics with respect to clinical and demographic factors. Fifty-two subjects with traumatic open fracture wounds (32 blunt and 20 penetrating injuries) were enrolled prospectively and sampled longitudinally from presentation to the emergency department (ED) and at each subsequent inpatient or outpatient encounter. Specimens were collected from both the wound center and adjacent skin. Culture-independent sequencing of the 16S ribosomal RNA gene was employed to identify and characterize microbiota. Upon presentation to the ED and time points immediately following, sample collection site (wound or adjacent skin) was the most defining feature discriminating microbial profiles. Microbial composition of adjacent skin and wound center converged over time. Mechanism of injury most strongly defined the microbiota after initial convergence. Further analysis controlling for race, gender, and age revealed that mechanism of injury remained a significant discriminating feature throughout the continuum of care. We conclude that the microbial communities associated with open fracture wounds are dynamic in nature until eventual convergence with the adjacent skin community during healing, with mechanism of injury as an important feature affecting both diversity and composition of the microbiota. A more complete understanding of the factors influencing microbial contamination and/or colonization in open fractures is a critical foundation for identifying markers indicative of outcome and deciphering their respective contributions to healing and/or complication.


Subject(s)
Bacteria/classification , Fractures, Open/microbiology , Microbiota/physiology , Skin/microbiology , Wound Healing/physiology , Wound Infection/microbiology , Adult , Aged , Bacteria/genetics , Colony Count, Microbial , Female , Fractures, Open/pathology , Humans , Longitudinal Studies , Male , Middle Aged , Pennsylvania , Prospective Studies , RNA, Ribosomal, 16S/genetics , Wound Infection/classification , Young Adult
8.
J Invest Dermatol ; 138(9): 1973-1981, 2018 09.
Article in English | MEDLINE | ID: mdl-29559344

ABSTRACT

Plaque psoriasis, a chronic inflammatory disease primarily affecting the skin, is thought to have a multifactorial etiology, including innate immune system dysregulation, environmental triggers, and genetic susceptibility. We sought to further understand the role of skin microbiota in psoriasis pathogenesis, as well as their response to therapy. We systematically analyzed dynamic microbiota colonizing psoriasis lesions and adjacent nonlesional skin in 114 patients prior to and during ustekinumab treatment in a phase 3b clinical trial. By sequencing the bacterial 16S ribosomal RNA gene from skin swab samples obtained at six anatomical sites, we identified minor, site-specific differences in microbial diversity and composition between pretreatment lesional and nonlesional skin. During therapy, microbial communities within lesional and nonlesional skin diverged, and body-site dispersion increased, reflecting microbial skin site-specificity. Microbiota demonstrated greater pretreatment heterogeneity in psoriatic lesions than in nonlesional skin, and variance increased as treatment progressed. Microbiota colonizing recurrent lesions did not overlap with pretreatment lesional microbiota, suggesting colonization patterns varied between initial and recurrent psoriatic lesions. While plaque psoriasis does not appear to be associated with specific microbes and/or microbial diversity, this large dataset provides insight into microbial variation associated with (i) disease in different body locations, (ii) initial versus recurrent lesions, and (iii) anti-IL12/23 therapy.


Subject(s)
Bacteria/genetics , Microbiota/drug effects , Psoriasis/drug therapy , Skin/microbiology , Ustekinumab/administration & dosage , Adolescent , Adult , Aged , Aged, 80 and over , Bacteria/isolation & purification , Cross-Sectional Studies , Dermatologic Agents/administration & dosage , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Humans , Male , Middle Aged , Psoriasis/metabolism , Psoriasis/microbiology , RNA, Bacterial/analysis , Retrospective Studies , Skin/pathology , Young Adult
9.
Microbiome ; 6(1): 20, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29378633

ABSTRACT

BACKGROUND: The skin harbors complex communities of resident microorganisms, yet little is known of their physiological roles and the molecular mechanisms that mediate cutaneous host-microbe interactions. Here, we profiled skin transcriptomes of mice reared in the presence and absence of microbiota to elucidate the range of pathways and functions modulated in the skin by the microbiota. RESULTS: A total of 2820 genes were differentially regulated in response to microbial colonization and were enriched in gene ontology (GO) terms related to the host-immune response and epidermal differentiation. Innate immune response genes and genes involved in cytokine activity were generally upregulated in response to microbiota and included genes encoding toll-like receptors, antimicrobial peptides, the complement cascade, and genes involved in IL-1 family cytokine signaling and homing of T cells. Our results also reveal a role for the microbiota in modulating epidermal differentiation and development, with differential expression of genes in the epidermal differentiation complex (EDC). Genes with correlated co-expression patterns were enriched in binding sites for the transcription factors Klf4, AP-1, and SP-1, all implicated as regulators of epidermal differentiation. Finally, we identified transcriptional signatures of microbial regulation common to both the skin and the gastrointestinal tract. CONCLUSIONS: With this foundational approach, we establish a critical resource for understanding the genome-wide implications of microbially mediated gene expression in the skin and emphasize prospective ways in which the microbiome contributes to skin health and disease.


Subject(s)
Gastrointestinal Tract/microbiology , Gene Expression Profiling/methods , Gene Regulatory Networks , Skin/microbiology , Animals , Cell Differentiation , Gastrointestinal Tract/immunology , Gene Expression Regulation , Host-Pathogen Interactions , Immunity, Innate , Kruppel-Like Factor 4 , Mice , Microbiota , Organ Specificity , Sequence Analysis, RNA/methods , Skin/immunology
10.
Pediatr Dermatol ; 34(6): 661-664, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29024079

ABSTRACT

BACKGROUND/OBJECTIVES: Acne, a common pediatric disease, tends to be more comedonal in preadolescents, whereas older individuals are more likely to have inflammatory lesions in addition to comedones. Thus the microbiome of preadolescents may be different. In this pilot study we aimed to characterize the preadolescent acne microbiome, compare the microbiome in preadolescents with and without acne, and investigate changes in the microbiome after topical treatment with benzoyl peroxide or a retinoid in a small cohort of preadolescents. METHODS: Participants were 7-10 years of age with (intervention group) or without (control group) acne and were recruited during routine outpatient dermatology visits. Baseline questionnaires, physical examination, and pore strip application were performed for all participants. Intervention group participants were randomized to receive topical therapy with benzoyl peroxide 5% gel or cream or tretinoin 0.025% cream. Participants with acne were followed up 8-10 weeks later and pore strip application was repeated. RESULTS: Preadolescents with acne were colonized with a greater diversity of cutaneous bacteria than controls and the most commonly identified bacterium was Streptococcus. The number of bacterial species and phylogenetic diversity decreased after treatment with benzoyl peroxide and tretinoin. CONCLUSION: The predominant bacteria in microbiome studies of adult acne is Propionibacterium, whereas in this pediatric population we saw a lot of Streptococcus bacteria. After treatment, the microbiomes of intervention group participants more closely resembled those of control group participants.


Subject(s)
Acne Vulgaris/drug therapy , Benzoyl Peroxide/administration & dosage , Dermatologic Agents/administration & dosage , Keratolytic Agents/administration & dosage , Microbiota/drug effects , Tretinoin/administration & dosage , Acne Vulgaris/microbiology , Administration, Topical , Child , Female , Humans , Male , Microbiota/genetics , Phylogeny , Pilot Projects , Prospective Studies , Skin/microbiology , Treatment Outcome
11.
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
12.
Article in English | MEDLINE | ID: mdl-28630195

ABSTRACT

The skin microbiome is a complex ecosystem with important implications for cutaneous health and disease. Topical antibiotics and antiseptics are often employed to preserve the balance of this population and inhibit colonization by more pathogenic bacteria. However, despite their widespread use, the impact of these interventions on broader microbial communities remains poorly understood. Here, we report the longitudinal effects of topical antibiotics and antiseptics on skin bacterial communities and their role in Staphylococcus aureus colonization resistance. In response to antibiotics, cutaneous populations exhibited an immediate shift in bacterial residents, an effect that persisted for multiple days posttreatment. By contrast, antiseptics elicited only minor changes to skin bacterial populations, with few changes to the underlying microbiota. While variable in scope, both antibiotics and antiseptics were found to decrease colonization by commensal Staphylococcus spp. by sequencing- and culture-based methods, an effect which was highly dependent on baseline levels of Staphylococcus Because Staphylococcus residents have been shown to compete with the skin pathogen S. aureus, we also tested whether treatment could influence S. aureus levels at the skin surface. We found that treated mice were more susceptible to exogenous association with S. aureus and that precolonization with the same Staphylococcus residents that were previously disrupted by treatment reduced S. aureus levels by over 100-fold. In all, the results of this study indicate that antimicrobial drugs can alter skin bacterial residents and that these alterations can have critical implications for cutaneous host defense.


Subject(s)
Anti-Bacterial Agents/pharmacology , Skin/microbiology , Staphylococcal Skin Infections/drug therapy , Staphylococcus aureus/drug effects , Administration, Cutaneous , Animals , Anti-Infective Agents, Local/pharmacology , Female , Mice , Microbiota/drug effects
13.
J Invest Dermatol ; 137(1): 237-244, 2017 01.
Article in English | MEDLINE | ID: mdl-27566400

ABSTRACT

Microbial burden of chronic wounds is believed to play an important role in impaired healing and the development of infection-related complications. However, clinical cultures have little predictive value of wound outcomes, and culture-independent studies have been limited by cross-sectional design and small cohort size. We systematically evaluated the temporal dynamics of the microbiota colonizing diabetic foot ulcers, a common and costly complication of diabetes, and its association with healing and clinical complications. Dirichlet multinomial mixture modeling, Markov chain analysis, and mixed-effect models were used to investigate shifts in the microbiota over time and their associations with healing. Here we show, to our knowledge, previously unreported temporal dynamics of the chronic wound microbiome. Microbiota community instability was associated with faster healing and improved outcomes. Diabetic foot ulcer microbiota were found to exist in one of four community types that experienced frequent and nonrandom transitions. Transition patterns and frequencies were associated with healing time. Exposure to systemic antibiotics destabilized the wound microbiota, rather than altering overall diversity or relative abundance of specific taxa. This study provides evidence that the dynamic wound microbiome is indicative of clinical outcomes and may be a valuable guide for personalized management and treatment of chronic wounds.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Diabetic Foot/drug therapy , Diabetic Foot/microbiology , Microbiota/drug effects , Wound Healing/physiology , Aged , Chronic Disease , Cohort Studies , Cross-Sectional Studies , Diabetic Foot/physiopathology , Disease Progression , Female , Humans , Longitudinal Studies , Male , Markov Chains , Middle Aged , Prognosis , Prospective Studies , Risk Assessment
14.
Clin Infect Dis ; 61(6): 892-9, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26206847

ABSTRACT

BACKGROUND: Klebsiella pneumoniae is a common colonizer of the gastrointestinal tract of humans, companion animals, and livestock. To better understand potential contributions of foodborne K. pneumoniae to human clinical infections, we compared K. pneumoniae isolates from retail meat products and human clinical specimens to assess their similarity based on antibiotic resistance, genetic relatedness, and virulence. METHODS: Klebsiella pneumoniae was isolated from retail meats from Flagstaff grocery stores in 2012 and from urine and blood specimens from Flagstaff Medical Center in 2011-2012. Isolates underwent antibiotic susceptibility testing and whole-genome sequencing. Genetic relatedness of the isolates was assessed using multilocus sequence typing and phylogenetic analyses. Extraintestinal virulence of several closely related meat-source and urine isolates was assessed using a murine sepsis model. RESULTS: Meat-source isolates were significantly more likely to be multidrug resistant and resistant to tetracycline and gentamicin than clinical isolates. Four sequence types occurred among both meat-source and clinical isolates. Phylogenetic analyses confirmed close relationships among meat-source and clinical isolates. Isolates from both sources showed similar virulence in the mouse sepsis model. CONCLUSIONS: Meat-source K. pneumoniae isolates were more likely than clinical isolates to be antibiotic resistant, which could reflect selective pressures from antibiotic use in food-animal production. The close genetic relatedness of meat-source and clinical isolates, coupled with similarities in virulence, suggest that the barriers to transmission between these 2 sources are low. Taken together, our results suggest that retail meat is a potential vehicle for transmitting virulent, antibiotic-resistant K. pneumoniae from food animals to humans.


Subject(s)
Klebsiella Infections/microbiology , Klebsiella pneumoniae/classification , Klebsiella pneumoniae/isolation & purification , Meat/microbiology , Urinary Tract Infections/microbiology , Adult , Aged , Aged, 80 and over , Animals , Anti-Bacterial Agents/pharmacology , Disease Models, Animal , Disease Transmission, Infectious , Genome, Bacterial , Genotype , Humans , Klebsiella Infections/epidemiology , Mice , Microbial Sensitivity Tests , Middle Aged , Molecular Epidemiology , Molecular Sequence Data , Multilocus Sequence Typing , Sepsis/epidemiology , Sepsis/microbiology , Sequence Analysis, DNA , Urinary Tract Infections/epidemiology , Virulence , Young Adult , Zoonoses/transmission
SELECTION OF CITATIONS
SEARCH DETAIL
...