90 results on '"Grice EA"'
Search Results
2. Commensal microbe regulation of skin cells in disease.
- Author
-
Gan Y, Zhang J, Qi F, Hu Z, Sweren E, Reddy SK, Chen L, Feng X, Grice EA, Garza LA, and Wang G
- Subjects
- Humans, Skin Diseases microbiology, Animals, Homeostasis, Host Microbial Interactions, Bacteria metabolism, Skin microbiology, Microbiota physiology, Symbiosis
- Abstract
Human skin is the host to various commensal microbes that constitute a substantial microbial community. The reciprocal communication between these microbial inhabitants and host cells upholds both the morphological and functional attributes of the skin layers, contributing indispensably to microenvironmental and tissue homeostasis. Thus, disruption of the skin barrier or imbalances in the microbial communities can exert profound effects on the behavior of host cells. This influence, mediated by the microbes themselves or their metabolites, manifests in diverse outcomes. In this review, we examine existing knowledge to provide insight into the nuanced behavior exhibited by the microbiota on skin cells in health and disease states. These interactions provide insight into potential cellular targets for future microbiota-based therapies to prevent and treat skin disease., Competing Interests: Declaration of interests L.A.G. has received grant support paid to his institution, Johns Hopkins University, from Sun Pharma Advanced Research Company (SPARC). This grant is to investigate intellectual property of which Johns Hopkins University is the owner. L.A.G. is one of several inventors and is under a licensing agreement with SPARC; this intellectual property has resulted in royalty payments to inventors. This grant and the royalty payments are not related to the research presented in this manuscript., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. Longitudinal evaluation of the cutaneous and rectal microbiota of German shepherd dogs with perianal fistulas undergoing therapy with ciclosporin and ketoconazole.
- Author
-
Cain CL, White E, Citron LE, Zheng Q, Morris DO, Grice EA, and Bradley CW 2nd
- Subjects
- Animals, Dogs, Male, Female, Longitudinal Studies, Rectum microbiology, Skin microbiology, Skin pathology, Microbiota drug effects, Cyclosporine therapeutic use, Cyclosporine administration & dosage, Dog Diseases drug therapy, Dog Diseases microbiology, Ketoconazole therapeutic use, Ketoconazole administration & dosage, Rectal Fistula veterinary, Rectal Fistula drug therapy, Rectal Fistula microbiology
- Abstract
Background: Perianal fistulas are painful ulcers or sinus tracts that disproportionately affect German shepherd dogs and are proposed as a spontaneous animal model of fistulising Crohn's disease., Objectives: To characterise the rectal and cutaneous microbiota in German shepherd dogs with perianal fistulas and to investigate longitudinal shifts with lesion resolution during immunomodulatory therapy., Animals: Eleven German shepherd dogs with perianal fistulas and 15 healthy German shepherd dogs., Materials and Methods: Affected dogs were evaluated and swabbed at three visits, 30 days apart, while undergoing treatment with ciclosporin and ketoconazole. Healthy German shepherd dogs were contemporaneously sampled. Sites included the rectum, perianal skin and axilla. The microbiome was evaluated following sequencing of the V4 hypervariable region of the 16S ribosomal RNA (rRNA) gene., Results: Alpha diversity was not significantly different between healthy and affected dogs at each of the three body sites (p > 0.5), yet rectal and perianal beta diversities from affected dogs differed significantly from those of healthy dogs at Day 0 (p = 0.004). Rectal and perianal relative abundance of Prevotella spp. increased and perianal Staphylococcus spp. relative abundance decreased in affected dogs over time, coincident with lesion resolution., Conclusions and Clinical Relevance: Changes in lesional cutaneous and rectal microbiota occur in German shepherd dogs with perianal fistulas and shift over time with lesion resolution during immunomodulatory therapy. Further investigations of the role of cutaneous and enteric microbiota in the pathogenesis of perianal fistulas, and whether manipulation of microbial populations may ameliorate disease, are needed., (© 2024 The Authors. Veterinary Dermatology published by John Wiley & Sons Ltd on behalf of ESVD and ACVD.)
- Published
- 2024
- Full Text
- View/download PDF
4. The wound microbiota: microbial mechanisms of impaired wound healing and infection.
- Author
-
Uberoi A, McCready-Vangi A, and Grice EA
- Subjects
- Humans, Skin microbiology, Biofilms growth & development, Animals, Wound Healing, Microbiota, Wound Infection microbiology
- Abstract
The skin barrier protects the human body from invasion by exogenous and pathogenic microorganisms. A breach in this barrier exposes the underlying tissue to microbial contamination, which can lead to infection, delayed healing, and further loss of tissue and organ integrity. Delayed wound healing and chronic wounds are associated with comorbidities, including diabetes, advanced age, immunosuppression and autoimmune disease. The wound microbiota can influence each stage of the multi-factorial repair process and influence the likelihood of an infection. Pathogens that commonly infect wounds, such as Staphylococcus aureus and Pseudomonas aeruginosa, express specialized virulence factors that facilitate adherence and invasion. Biofilm formation and other polymicrobial interactions contribute to host immunity evasion and resistance to antimicrobial therapies. Anaerobic organisms, fungal and viral pathogens, and emerging drug-resistant microorganisms present unique challenges for diagnosis and therapy. In this Review, we explore the current understanding of how microorganisms present in wounds impact the process of skin repair and lead to infection through their actions on the host and the other microbial wound inhabitants., (© 2024. Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
5. Alcaligenes faecalis corrects aberrant matrix metalloproteinase expression to promote reepithelialization of diabetic wounds.
- Author
-
White EK, Uberoi A, Pan JT, Ort JT, Campbell AE, Murga-Garrido SM, Harris JC, Bhanap P, Wei M, Robles NY, Gardner SE, and Grice EA
- Subjects
- Animals, Humans, Diabetic Foot microbiology, Diabetic Foot pathology, Diabetic Foot metabolism, Mice, Re-Epithelialization, Male, Alcaligenes faecalis metabolism, Wound Healing, Keratinocytes metabolism, Keratinocytes microbiology, Matrix Metalloproteinases metabolism, Matrix Metalloproteinases genetics
- Abstract
Chronic wounds are a common and costly complication of diabetes, where multifactorial defects contribute to dysregulated skin repair, inflammation, tissue damage, and infection. We previously showed that aspects of the diabetic foot ulcer microbiota were correlated with poor healing outcomes, but many microbial species recovered remain uninvestigated with respect to wound healing. Here, we focused on Alcaligenes faecalis , a Gram-negative bacterium that is frequently recovered from chronic wounds but rarely causes infection. Treatment of diabetic wounds with A. faecalis accelerated healing during early stages. We investigated the underlying mechanisms and found that A. faecalis treatment promotes reepithelialization of diabetic keratinocytes, a process that is necessary for healing but deficient in chronic wounds. Overexpression of matrix metalloproteinases in diabetes contributes to failed epithelialization, and we found that A. faecalis treatment balances this overexpression to allow proper healing. This work uncovers a mechanism of bacterial-driven wound repair and provides a foundation for the development of microbiota-based wound interventions.
- Published
- 2024
- Full Text
- View/download PDF
6. Laser-Capture Microdissection-Based RNA Sequencing for Profiling Mouse and Human Sebaceous Gland Transcriptomes.
- Author
-
Harris JC, Prouty SM, Nelson MA, Sung DC, Nelson AM, Seykora JT, Kambayashi T, and Grice EA
- Subjects
- Humans, Mice, Animals, Gene Expression Profiling methods, Sebaceous Glands metabolism, Sebaceous Glands pathology, Transcriptome, Laser Capture Microdissection methods, Sequence Analysis, RNA methods
- Published
- 2024
- Full Text
- View/download PDF
7. The microbiota and T cells non-genetically modulate inherited phenotypes transgenerationally.
- Author
-
Harris JC, Trigg NA, Goshu B, Yokoyama Y, Dohnalová L, White EK, Harman A, Murga-Garrido SM, Ting-Chun Pan J, Bhanap P, Thaiss CA, Grice EA, Conine CC, and Kambayashi T
- Subjects
- Animals, Mice, Male, Female, Mice, Inbred C57BL, Phenotype, T-Lymphocytes immunology, T-Lymphocytes metabolism, Microbiota
- Abstract
The host-microbiota relationship has evolved to shape mammalian physiology, including immunity, metabolism, and development. Germ-free models are widely used to study microbial effects on host processes such as immunity. Here, we find that both germ-free and T cell-deficient mice exhibit a robust sebum secretion defect persisting across multiple generations despite microbial colonization and T cell repletion. These phenotypes are inherited by progeny conceived during in vitro fertilization using germ-free sperm and eggs, demonstrating that non-genetic information in the gametes is required for microbial-dependent phenotypic transmission. Accordingly, gene expression in early embryos derived from gametes from germ-free or T cell-deficient mice is strikingly and similarly altered. Our findings demonstrate that microbial- and immune-dependent regulation of non-genetic information in the gametes can transmit inherited phenotypes transgenerationally in mice. This mechanism could rapidly generate phenotypic diversity to enhance host adaptation to environmental perturbations., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. An exploration of mechanisms underlying Desemzia incerta colonization resistance to methicillin-resistant Staphylococcus aureus on the skin.
- Author
-
Wei M, Knight SAB, Fazelinia H, Spruce L, Roof J, Chu E, Kim DY, Bhanap P, Walsh J, Flowers L, Zhu J, and Grice EA
- Subjects
- Humans, Swine, Animals, Mice, Staphylococcus aureus, Proteomics, Methicillin-Resistant Staphylococcus aureus, Anti-Infective Agents, Carnobacteriaceae
- Abstract
Colonization of human skin and nares by methicillin-resistant Staphylococcus aureus (MRSA) leads to the community spread of MRSA. This spread is exacerbated by the transfer of MRSA between humans and livestock, particularly swine. Here, we capitalized on the shared features between human and porcine skin, including shared MRSA colonization, to study novel bacterial mediators of MRSA colonization resistance. We focused on the poorly studied bacterial species Desemzia incerta , which we found to exert antimicrobial activity through a secreted product and exhibited colonization resistance against MRSA in an in vivo murine skin model. Using parallel genomic and biochemical investigation, we discovered that D. incerta secretes an antimicrobial protein. Sequential protein purification and proteomics analysis identified 24 candidate inhibitory proteins, including a promising peptidoglycan hydrolase candidate. Aided by transcriptional analysis of D. incerta and MRSA cocultures, we found that exposure to D. incerta leads to decreased MRSA biofilm production. These results emphasize the value of exploring microbial communities across a spectrum of hosts, which can lead to novel therapeutic agents as well as an increased understanding of microbial competition.IMPORTANCEMethicillin-resistant Staphylococcus aureus (MRSA) causes a significant healthcare burden and can be spread to the human population via livestock transmission. Members of the skin microbiome can prevent MRSA colonization via a poorly understood phenomenon known as colonization resistance. Here, we studied the colonization resistance of S. aureus by bacterial inhibitors previously identified from a porcine skin model. We identify a pig skin commensal, Desemzia incerta , that reduced MRSA colonization in a murine model. We employ a combination of genomic, proteomic, and transcriptomic analyses to explore the mechanisms of inhibition between D. incerta and S. aureus . We identify 24 candidate antimicrobial proteins secreted by D. incerta that could be responsible for its antimicrobial activity. We also find that exposure to D. incerta leads to decreased S. aureus biofilm formation. These findings show that the livestock transmission of MRSA can be exploited to uncover novel mechanisms of MRSA colonization resistance., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
9. Pregnant Females with Atopic Dermatitis Are More Likely to be Colonized with Group B Streptococci.
- Author
-
Margolis DJ, Chiesa Fuxench ZC, Hoffstad OJ, Grice EA, and Mitra N
- Subjects
- Pregnancy, Female, Humans, Skin, Staphylococcus aureus, Dermatitis, Atopic epidemiology
- Published
- 2024
- Full Text
- View/download PDF
10. Regulatory T cells control Staphylococcus aureus and disease severity of cutaneous leishmaniasis.
- Author
-
Singh TP, Farias Amorim C, Lovins VM, Bradley CW, Carvalho LP, Carvalho EM, Grice EA, and Scott P
- Subjects
- Humans, Animals, Mice, Staphylococcus aureus, Interleukin-17, T-Lymphocytes, Regulatory, Patient Acuity, Forkhead Transcription Factors, Leishmaniasis, Cutaneous, Staphylococcal Infections
- Abstract
Cutaneous leishmaniasis causes alterations in the skin microbiota, leading to pathologic immune responses and delayed healing. However, it is not known how these microbiota-driven immune responses are regulated. Here, we report that depletion of Foxp3+ regulatory T cells (Tregs) in Staphylococcus aureus-colonized mice resulted in less IL-17 and an IFN-γ-dependent skin inflammation with impaired S. aureus immunity. Similarly, reducing Tregs in S. aureus-colonized and Leishmania braziliensis-infected mice increased IFN-γ, S. aureus, and disease severity. Importantly, analysis of lesions from L. braziliensis patients revealed that low FOXP3 gene expression is associated with high IFNG expression, S. aureus burden, and delayed lesion resolution compared to patients with high FOXP3 expression. Thus, we found a critical role for Tregs in regulating the balance between IL-17 and IFN-γ in the skin, which influences both bacterial burden and disease. These results have clinical ramifications for cutaneous leishmaniasis and other skin diseases associated with a dysregulated microbiome when Tregs are limited or dysfunctional., (© 2023 Singh et al.)
- Published
- 2023
- Full Text
- View/download PDF
11. Theranostic gold-in-gold cage nanoparticles enable photothermal ablation and photoacoustic imaging in biofilm-associated infection models.
- Author
-
Hajfathalian M, de Vries CR, Hsu JC, Amirshaghaghi A, Dong YC, Ren Z, Liu Y, Huang Y, Li Y, Knight SA, Jonnalagadda P, Zlitni A, Grice EA, Bollyky PL, Koo H, and Cormode DP
- Subjects
- Animals, Mice, Anti-Bacterial Agents, Biofilms, Gold pharmacology, Gold chemistry, Precision Medicine, Nanoparticles chemistry, Photoacoustic Techniques, Wound Infection
- Abstract
Biofilms are structured communities of microbial cells embedded in a self-produced matrix of extracellular polymeric substances. Biofilms are associated with many health issues in humans, including chronic wound infections and tooth decay. Current antimicrobials are often incapable of disrupting the polymeric biofilm matrix and reaching the bacteria within. Alternative approaches are needed. Here, we described a complex structure of a dextran-coated gold-in-gold cage nanoparticle that enabled photoacoustic and photothermal properties for biofilm detection and treatment. Activation of these nanoparticles with a near infrared laser could selectively detect and kill biofilm bacteria with precise spatial control and in a short timeframe. We observed a strong biocidal effect against both Streptococcus mutans and Staphylococcus aureus biofilms in mouse models of oral plaque and wound infections, respectively. These effects were over 100 times greater than those seen with chlorhexidine, a conventional antimicrobial agent. Moreover, this approach did not adversely affect surrounding tissues. We concluded that photothermal ablation using theranostic nanoparticles is a rapid, precise, and nontoxic method to detect and treat biofilm-associated infections.
- Published
- 2023
- Full Text
- View/download PDF
12. Variable staphyloxanthin production by Staphylococcus aureus drives strain-dependent effects on diabetic wound-healing outcomes.
- Author
-
Campbell AE, McCready-Vangi AR, Uberoi A, Murga-Garrido SM, Lovins VM, White EK, Pan JT, Knight SAB, Morgenstern AR, Bianco C, Planet PJ, Gardner SE, and Grice EA
- Subjects
- Animals, Mice, Staphylococcus aureus metabolism, Wound Healing, Staphylococcal Infections microbiology, Diabetes Mellitus
- Abstract
Strain-level variation in Staphylococcus aureus is a factor that contributes to disease burden and clinical outcomes in skin disorders and chronic wounds. However, the microbial mechanisms that drive these variable host responses are poorly understood. To identify mechanisms underlying strain-specific outcomes, we perform high-throughput phenotyping screens on S. aureus isolates cultured from diabetic foot ulcers. Isolates from non-healing wounds produce more staphyloxanthin, a cell membrane pigment. In murine diabetic wounds, staphyloxanthin-producing isolates delay wound closure significantly compared with staphyloxanthin-deficient isolates. Staphyloxanthin promotes resistance to oxidative stress and enhances bacterial survival in neutrophils. Comparative genomic and transcriptomic analysis of genetically similar clinical isolates with disparate staphyloxanthin phenotypes reveals a mutation in the sigma B operon, resulting in marked differences in stress response gene expression. Our work illustrates a framework to identify traits that underlie strain-level variation in disease burden and suggests more precise targets for therapeutic intervention in S. aureus-positive wounds., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
13. Multiomic profiling of cutaneous leishmaniasis infections reveals microbiota-driven mechanisms underlying disease severity.
- Author
-
Farias Amorim C, Lovins VM, Singh TP, Novais FO, Harris JC, Lago AS, Carvalho LP, Carvalho EM, Beiting DP, Scott P, and Grice EA
- Subjects
- Humans, Mice, Animals, Staphylococcus aureus, Multiomics, Inflammation, Bacteria, Patient Acuity, Leishmaniasis, Cutaneous, Microbiota
- Abstract
Leishmania braziliensis is a parasitic infection that can result in inflammation and skin injury with highly variable and unpredictable clinical outcomes. Here, we investigated the potential impact of microbiota on infection-induced inflammatory responses and disease resolution by conducting an integrated analysis of the skin microbiome and host transcriptome on a cohort of 62 patients infected with L. braziliensis . We found that overall bacterial burden and microbiome configurations dominated with Staphylococcus spp. were associated with delayed healing and enhanced inflammatory responses, especially by IL-1 family members. Quantification of host and bacterial transcripts on human lesions revealed that high lesional S. aureus transcript abundance was associated with delayed healing and increased expression of IL-1β. This cytokine was critical for modulating disease outcomes in L. braziliensis -infected mice colonized with S. aureus , given that its neutralization reduced pathology and inflammation. These results highlight how the human microbiome can shape disease outcomes in cutaneous leishmaniasis and suggest pathways toward host-directed therapies to mitigate the inflammatory consequences.
- Published
- 2023
- Full Text
- View/download PDF
14. An exploration of mechanisms underlying Desemzia incerta colonization resistance to methicillin-resistant Staphylococcus aureus on the skin.
- Author
-
Wei M, Knight SA, Fazelinia H, Spruce L, Roof J, Chu E, Walsh J, Flowers L, Kim DY, Zhu J, and Grice EA
- Abstract
Colonization of human skin and nares by methicillin-resistant Staphylococcus aureus (MRSA) leads to community spread of MRSA. This spread is exacerbated by transfer of MRSA between humans and livestock, particularly swine. Here we capitalized on the shared features between human and porcine skin, including shared MRSA colonization, to study novel bacterial mediators of MRSA colonization resistance. We focused on the poorly studied bacterial species Desemzia incerta , which we found to exert antimicrobial activity through a secreted product and exhibited colonization resistance against MRSA in an in vivo murine skin model. Using parallel genomic and biochemical investigation, we discovered that D. incerta secretes an antimicrobial protein. Sequential protein purification and proteomics analysis identified 24 candidate inhibitory proteins, including a promising peptidoglycan hydrolase candidate. Aided by transcriptional analysis of D. incerta and MRSA cocultures, we found that exposure to D. incerta leads to decreased MRSA biofilm production. These results emphasize the value in exploring microbial communities across a spectrum of hosts, which can lead to novel therapeutic agents as well as increased understanding of microbial competition.
- Published
- 2023
- Full Text
- View/download PDF
15. Ligand Activation of the Aryl Hydrocarbon Receptor Upregulates Epidermal Uridine Diphosphate Glucose Ceramide Glucosyltransferase and Glucosylceramides.
- Author
-
Sutter CH, Azim S, Wang A, Bhuju J, Simpson AS, Uberoi A, Grice EA, and Sutter TR
- Subjects
- Animals, Mice, Humans, Uridine Diphosphate Glucose, Receptors, Aryl Hydrocarbon genetics, Receptors, Aryl Hydrocarbon metabolism, Ligands, RNA, Glucosylceramides metabolism, Polychlorinated Dibenzodioxins
- Abstract
Ligand activation of the aryl hydrocarbon receptor (AHR) accelerates keratinocyte differentiation and the formation of the epidermal permeability barrier. Several classes of lipids, including ceramides, are critical to the epidermal permeability barrier. In normal human epidermal keratinocytes, the AHR ligand, 2,3,7,8-tetrachlorodibenzo-p-dioxin, increased RNA levels of ceramide metabolism and transport genes: uridine diphosphate glucose ceramide glucosyltransferase (UGCG), ABCA12, GBA1, and SMPD1. Levels of abundant skin ceramides were also increased by 2,3,7,8-tetrachlorodibenzo-p-dioxin. These included the metabolites synthesized by UGCG, glucosylceramides, and acyl glucosylceramides. Chromatin immunoprecipitation-sequence analysis and luciferase reporter assays identified UGCG as a direct AHR target. The AHR antagonist, GNF351, inhibited the 2,3,7,8-tetrachlorodibenzo-p-dioxin-mediated RNA and transcriptional increases. Tapinarof, an AHR ligand approved for the treatment of psoriasis, increased UGCG RNA, protein, and its lipid metabolites hexosylceramides as well as increased the RNA expression of ABCA12, GBA1, and SMPD1. In Ahr-null mice, Ugcg RNA and hexosylceramides were lower than those in the wild type. These results indicate that the AHR regulates the expression of UGCG, a ceramide-metabolizing enzyme required for ceramide trafficking, keratinocyte differentiation, and epidermal permeability barrier formation., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
16. Harnessing diversity and antagonism within the pig skin microbiota to identify novel mediators of colonization resistance to methicillin-resistant Staphylococcus aureus .
- Author
-
Wei M, Flowers L, Knight SAB, Zheng Q, Murga-Garrido S, Uberoi A, Pan JT, Walsh J, Schroeder E, Chu EW, Campbell A, Shin D, Bradley CW, Duran-Struuck R, and Grice EA
- Subjects
- Animals, Swine, Mice, Staphylococcus aureus genetics, Nasal Cavity, Methicillin-Resistant Staphylococcus aureus genetics, Staphylococcal Infections prevention & control, Staphylococcal Infections veterinary, Microbiota
- Abstract
The microbiota mediate multiple aspects of skin barrier function, including colonization resistance to pathogens such as Staphylococcus aureus . The endogenous skin microbiota limits S. aureus colonization via competition and direct inhibition. Novel mechanisms of colonization resistance are promising therapeutic targets for drug-resistant infections, such as those caused by methicillin-resistant S. aureus (MRSA). Here, we developed and characterized a swine model of topical microbiome perturbation and MRSA colonization. As in other model systems, topical antimicrobial treatment had a little discernable effect on community diversity though the overall microbial load was sensitive to multiple types of intervention, including swabbing. In parallel, we established a porcine skin culture collection and screened 7,700 isolates for MRSA inhibition. Using genomic and phenotypic criteria, we curated three isolates to investigate whether prophylactic colonization would inhibit MRSA colonization in vivo . The three-member consortium together, but not individually, provided protection against MRSA colonization, suggesting cooperation and/or synergy among the strains. Inhibitory isolates were represented across all major phyla of the pig skin microbiota and did not have a strong preference for inhibiting closely related species, suggesting that relatedness is not a condition of antagonism. These findings reveal the porcine skin as an underexplored reservoir of skin commensal species with the potential to prevent MRSA colonization and infection. IMPORTANCE The skin microbiota is protective against pathogens or opportunists such as S. aureus , the most common cause of skin and soft tissue infections. S. aureus can colonize normal skin and nasal passages, and colonization is a risk factor for infection, especially on breach of the skin barrier. Here, we established a pig model to study the competitive mechanisms of the skin microbiota and their role in preventing colonization by MRSA. This drug-resistant strain is also a livestock pathogen, and swine herds can be reservoirs of MRSA carriage. From 7,700 cultured skin isolates, we identified 37 unique species across three phyla that inhibited MRSA. A synthetic community of three inhibitory isolates provided protection together, but not individually, in vivo in a murine model of MRSA colonization. These findings suggest that antagonism is widespread in the pig skin microbiota, and these competitive interactions may be exploited to prevent MRSA colonization., Competing Interests: The authors declare no conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
17. Wound microbiota-mediated correction of matrix metalloproteinase expression promotes re-epithelialization of diabetic wounds.
- Author
-
White EK, Uberoi A, Pan JT, Ort JT, Campbell AE, Murga-Garrido SM, Harris JC, Bhanap P, Wei M, Robles NY, Gardner SE, and Grice EA
- Abstract
Chronic wounds are a common and costly complication of diabetes, where multifactorial defects contribute to dysregulated skin repair, inflammation, tissue damage, and infection. We previously showed that aspects of the diabetic foot ulcer microbiota were correlated with poor healing outcomes, but many microbial species recovered remain uninvestigated with respect to wound healing. Here we focused on Alcaligenes faecalis , a Gram-negative bacterium that is frequently recovered from chronic wounds but rarely causes infection. Treatment of diabetic wounds with A. faecalis accelerated healing during early stages. We investigated the underlying mechanisms and found that A. faecalis treatment promotes re-epithelialization of diabetic keratinocytes, a process which is necessary for healing but deficient in chronic wounds. Overexpression of matrix metalloproteinases in diabetes contributes to failed epithelialization, and we found that A. faecalis treatment balances this overexpression to allow proper healing. This work uncovers a mechanism of bacterial-driven wound repair and provides a foundation for the development of microbiota-based wound interventions.
- Published
- 2023
- Full Text
- View/download PDF
18. Involucrin Modulates Vitamin D Receptor Activity in the Epidermis.
- Author
-
Schmidt AD, Miciano C, Zheng Q, Mathyer ME, Grice EA, and de Guzman Strong C
- Subjects
- Mice, Humans, Animals, Skin metabolism, Keratinocytes metabolism, Vitamin D pharmacology, Vitamin D metabolism, Inflammation metabolism, Receptors, Calcitriol metabolism, Epidermis metabolism
- Abstract
Terminally differentiated keratinocytes are critical for epidermal function and are surrounded by involucrin (IVL). Increased IVL expression is associated with a near-selective sweep in European populations compared with those in Africa. This positive selection for increased IVL in the epidermis identifies human adaptation outside of Africa. The functional significance is unclear. We hypothesize that IVL modulates the environmentally sensitive vitamin D receptor (VDR) in the epidermis. We investigated VDR activity in Ivl
‒/‒ and wild-type mice using vitamin D agonist (MC903) treatment and comprehensively determined the inflammatory response using single-cell RNA sequencing and associated skin microbiome changes using 16S bacterial phylotyping. VDR activity and target gene expression were reduced in Ivl‒/‒ mouse skin, with decreased MC903-mediated skin inflammation and significant reductions in CD4+ T cells, basophils, macrophages, monocytes, and type II basal keratinocytes and an increase in suprabasal keratinocytes. Coinciding with the dampened MC903-mediated inflammation, the skin microbiota of Ivl‒/‒ mice was more stable than that of the wild-type mice, which exhibited an MC903-responsive increase in Bacteroidetes and a decrease in Firmicutes. Together, our studies in Ivl‒/‒ mice identify a functional role for IVL to positively impact VDR activity and suggest an emerging IVL/VDR paradigm for adaptation in the human epidermis., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
19. Crosstalk between skin microbiota and immune system in health and disease.
- Author
-
Liu Q, Ranallo R, Rios C, Grice EA, Moon K, and Gallo RL
- Subjects
- Skin, Immune System, Microbiota
- Published
- 2023
- Full Text
- View/download PDF
20. The Wound Microbiome.
- Author
-
White EK and Grice EA
- Subjects
- Wound Healing, Bacteria, Fungi, Microbiota
- Abstract
Breach of the skin barrier and subsequent wound healing occur in the context of microbial communities of bacteria, fungi, and viruses. These polymicrobial communities are dynamic and important components of the wound environment and are associated with differential healing outcomes. Here, we highlight both culture-dependent and -independent methods that have furthered our understanding of the wound microbiome. We discuss common themes that have developed from such studies about the microbial inhabitants of diverse wound types. We additionally explore the wide range of microbial mechanisms that influence healing, from invading pathogens to beneficial commensals. These insights can be leveraged to better predict healing outcomes and derive novel microbial-based therapies for chronic wounds., (Copyright © 2023 Cold Spring Harbor Laboratory Press; all rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
21. The microbiota and immune system non-genetically affect offspring phenotypes transgenerationally.
- Author
-
Harris JC, Trigg NA, Goshu B, Yokoyama Y, Dohnalová L, White EK, Harman A, Thaiss CA, Grice EA, Conine CC, and Kambayashi T
- Abstract
The host-microbiota relationship has evolved to shape mammalian processes, including immunity, metabolism, and development
1-3 . Host phenotypes change in direct response to microbial exposures by the individual. Here we show that the microbiota induces phenotypic change not only in the individual but also in their succeeding generations of progeny. We found that germ-free mice exhibit a robust sebum secretion defect and transcriptional changes in various organs, persisting across multiple generations despite microbial colonization and breeding with conventional mice. Host-microbe interactions could be involved in this process, since T cell-deficient mice, which display defective sebum secretion4 , also transgenerationally transmit their phenotype to progeny. These phenotypes are inherited by progeny conceived during in vitro fertilization using germ-free sperm and eggs, demonstrating that epigenetic information in the gametes is required for phenotypic transmission. Accordingly, small non-coding RNAs that can regulate embryonic gene expression5 were strikingly and similarly altered in gametes of germ-free and T cell-deficient mice. Thus, we have uncovered a novel mechanism whereby the microbiota and immune system induce phenotypic changes in successive generations of offspring. This epigenetic form of inheritance could be advantageous for host adaptation to environmental perturbation, where phenotypic diversity can be introduced more rapidly than by genetic mutation.- Published
- 2023
- Full Text
- View/download PDF
22. The skin microbiome enhances disease through IL-1b and delays healing in cutaneous leishmaniasis patients.
- Author
-
Farias Amorim C, Lovins VM, Singh TP, Novais FO, Harris JC, Lago AS, Carvalho LP, Carvalho EM, Beiting DP, Scott P, and Grice EA
- Abstract
Leishmania braziliensis infection results in inflammation and skin injury, with highly variable and unpredictable clinical outcomes. Here, we investigated the potential impact of microbiota on infection-induced inflammatory responses and disease resolution by conducting an integrated analysis of the skin microbiome and host transcriptome on a cohort of 62 L. braziliensis -infected patients. We found that overall bacterial burden and microbiome configurations dominated with Staphylococcus spp. were associated with delayed healing and enhanced inflammatory responses, especially by IL-1 family members. Dual RNA-seq of human lesions revealed that high lesional S. aureus transcript abundance was associated with delayed healing and increased expression of IL-1β. This cytokine was critical for modulating disease outcome in L. braziliensis -infected mice colonized with S. aureus , as its neutralization reduced pathology and inflammation. These results implicate the microbiome in cutaneous leishmaniasis disease outcomes in humans and suggest host-directed therapies to mitigate the inflammatory consequences.
- Published
- 2023
- Full Text
- View/download PDF
23. Commensal microbiome promotes hair follicle regeneration by inducing keratinocyte HIF-1α signaling and glutamine metabolism.
- Author
-
Wang G, Sweren E, Andrews W, Li Y, Chen J, Xue Y, Wier E, Alphonse MP, Luo L, Miao Y, Chen R, Zeng D, Lee S, Li A, Dare E, Kim D, Archer NK, Reddy SK, Resar L, Hu Z, Grice EA, Kane MA, and Garza LA
- Subjects
- Animals, Humans, Mice, Keratinocytes, Regeneration, Skin metabolism, Wound Healing, Microbiota, Glutamine metabolism, Hair Follicle
- Abstract
Tissue injury induces metabolic changes in stem cells, which likely modulate regeneration. Using a model of organ regeneration called wound-induced hair follicle neogenesis (WIHN), we identified skin-resident bacteria as key modulators of keratinocyte metabolism, demonstrating a positive correlation between bacterial load, glutamine metabolism, and regeneration. Specifically, through comprehensive multiomic analysis and single-cell RNA sequencing in murine skin, we show that bacterially induced hypoxia drives increased glutamine metabolism in keratinocytes with attendant enhancement of skin and hair follicle regeneration. In human skin wounds, topical broad-spectrum antibiotics inhibit glutamine production and are partially responsible for reduced healing. These findings reveal a conserved and coherent physiologic context in which bacterially induced metabolic changes improve the tolerance of stem cells to damage and enhance regenerative capacity. This unexpected proregenerative modulation of metabolism by the skin microbiome in both mice and humans suggests important methods for enhancing regeneration after injury.
- Published
- 2023
- Full Text
- View/download PDF
24. Microbiota and maintenance of skin barrier function.
- Author
-
Harris-Tryon TA and Grice EA
- Subjects
- Epithelium immunology, Epithelium microbiology, Hair Follicle immunology, Hair Follicle microbiology, Humans, Skin Diseases immunology, Skin Diseases therapy, Wound Healing immunology, Host Microbial Interactions immunology, Microbiota, Skin immunology, Skin microbiology
- Abstract
Human skin forms a protective barrier against the external environment and is our first line of defense against toxic, solar, and pathogenic insults. Our skin also defines our outward appearance, protects our internal tissues and organs, acts as a sensory interface, and prevents dehydration. Crucial to the skin's barrier function is the colonizing microbiota, which provides protection against pathogens, tunes immune responses, and fortifies the epithelium. Here we highlight recent advances in our understanding of how the microbiota mediates multiple facets of skin barrier function. We discuss recent insights into pathological host-microbiota interactions and implications for disorders of the skin and distant organs. Finally, we examine how microbiota-based mechanisms can be targeted to prevent or manage skin disorders and impaired wound healing.
- Published
- 2022
- Full Text
- View/download PDF
25. Microbiota instruct IL-17A-producing innate lymphoid cells to promote skin inflammation in cutaneous leishmaniasis.
- Author
-
Singh TP, Carvalho AM, Sacramento LA, Grice EA, and Scott P
- Subjects
- Animals, Dermatitis immunology, Dermatitis microbiology, Immunity, Innate immunology, Leishmaniasis, Cutaneous microbiology, Mice, Dendritic Cells immunology, Interleukin-17 immunology, Leishmaniasis, Cutaneous immunology, Lymphocytes immunology, Skin microbiology
- Abstract
Innate lymphoid cells (ILCs) comprise a heterogeneous population of immune cells that maintain barrier function and can initiate a protective or pathological immune response upon infection. Here we show the involvement of IL-17A-producing ILCs in microbiota-driven immunopathology in cutaneous leishmaniasis. IL-17A-producing ILCs were RORγt+ and were enriched in Leishmania major infected skin, and topical colonization with Staphylococcus epidermidis before L. major infection exacerbated the skin inflammatory responses and IL-17A-producing RORγt+ ILC accumulation without impacting type 1 immune responses. IL-17A responses in ILCs were directed by Batf3 dependent CD103+ dendritic cells and IL-23. Moreover, experiments using Rag1-/- mice established that IL-17A+ ILCs were sufficient in driving the inflammatory responses as depletion of ILCs or neutralization of IL-17A diminished the microbiota mediated immunopathology. Taken together, this study indicates that the skin microbiota promotes RORγt+ IL-17A-producing ILCs, which augment the skin inflammation in cutaneous leishmaniasis., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
26. Skin microbiome and antimicrobial peptides.
- Author
-
Zeeuwen PLJM and Grice EA
- Subjects
- Humans, Antimicrobial Peptides immunology, Microbiota immunology, Skin immunology, Skin microbiology
- Published
- 2021
- Full Text
- View/download PDF
27. Insights into the skin microbiome of sickle cell disease leg ulcers.
- Author
-
Byeon J, Blizinsky KD, Persaud A, Findley K, Lee JJ, Buscetta AJ, You S, Bittinger K, Minniti CP, Bonham VL, and Grice EA
- Subjects
- Cross-Sectional Studies, Humans, Wound Healing, Anemia, Sickle Cell, Leg Ulcer, Microbiota
- Abstract
Leg ulcers are estimated to occur in 1%-10% of North American patients with sickle cell disease (SCD). Their pathophysiology remains poorly defined, but as with other chronic wounds, it is hypothesised that the microbial milieu, or microbiome, contributes to their healing and clinical outcomes. This study utilises 16S ribosomal RNA (rRNA) gene sequencing to describe, for the first time, the microbiome of the SCD leg ulcer and its association with clinical factors. In a cross-sectional analysis of 42 ulcers, we recovered microbial profiles similar to other chronic wounds in the predominance of anaerobic bacteria and opportunistic pathogens including Staphylococcus, Corynebacterium, and Finegoldia. Ulcers separated into two clusters: one defined by predominance of Staphylococcus and smaller surface area, and the other displaying a greater diversity of taxa and larger surface area. We also find that the relative abundance of Porphyromonas is negatively associated with haemoglobin levels, a key clinical severity indicator for SCD, and that Finegoldia relative abundance is negatively associated with CD19+ B cell count. Finally, ratios of Corynebacterium:Lactobacillus and Staphylococcus:Lactobacillus are elevated in the intact skin of individuals with a history of SCD leg ulcers, while the ratio of Lactobacillus:Bacillus is elevated in that of individuals without a history of ulcers. Investigations of the skin microbiome in relation to SCD ulcer pathophysiology can inform clinical guidelines for this poorly understood chronic wound, as well as enhance broader understanding about the role of the skin microbiome in delayed wound healing., (© 2021 The Authors. Wound Repair and Regeneration published by Wiley Periodicals LLC on behalf of The Wound Healing Society.)
- Published
- 2021
- Full Text
- View/download PDF
28. Cutaneous Effects of In Utero and Lactational Exposure of C57BL/6J Mice to 2,3,7,8-Tetrachlorodibenzo- p -dioxin.
- Author
-
Bhuju J, Olesen KM, Muenyi CS, Patel TS, Read RW, Thompson L, Skalli O, Zheng Q, Grice EA, Sutter CH, and Sutter TR
- Abstract
To determine the cutaneous effects of in utero and lactational exposure to the AHR ligand 2,3,7,8-tetrachlorodibenzo- p -dioxin (TCDD), pregnant C57BL/6J mice were exposed by gavage to a vehicle or 5 μg TCDD/kg body weight at embryonic day 12 and epidermal barrier formation and function were studied in their offspring from postnatal day 1 (P1) through adulthood. TCDD-exposed pups were born with acanthosis. This effect was AHR-dependent and subsided by P6 with no evidence of subsequent inflammatory dermatitis. The challenge of adult mice with MC903 showed similar inflammatory responses in control and treated animals, indicating no long-term immunosuppression to this chemical. Chloracne-like sebaceous gland hypoplasia and cyst formation were observed in TCDD-exposed P21 mice, with concomitant microbiome dysbiosis. These effects were reversed by P35. CYP1A1 and CYP1B1 expression in the skin was increased in the exposed mice until P21, then declined. Both CYP proteins co-localized with LRIG1-expressing progenitor cells at the infundibulum. CYP1B1 protein also co-localized with a second stem cell niche in the isthmus. These results indicate that this exposure to TCDD causes a chloracne-like effect without inflammation. Transient activation of the AhR, due to the shorter half-life of TCDD in mice, likely contributes to the reversibility of these effects.
- Published
- 2021
- Full Text
- View/download PDF
29. Commensal microbiota regulates skin barrier function and repair via signaling through the aryl hydrocarbon receptor.
- Author
-
Uberoi A, Bartow-McKenney C, Zheng Q, Flowers L, Campbell A, Knight SAB, Chan N, Wei M, Lovins V, Bugayev J, Horwinski J, Bradley C, Meyer J, Crumrine D, Sutter CH, Elias P, Mauldin E, Sutter TR, and Grice EA
- Subjects
- 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, Microbiota physiology, Receptors, Aryl Hydrocarbon metabolism, Signal Transduction, Skin microbiology
- 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., Competing Interests: Declaration of interests The authors have no competing interests to declare., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
30. Microbial Sharing between Pediatric Patients and Therapy Dogs during Hospital Animal-Assisted Intervention Programs.
- Author
-
Dalton KR, Ruble K, Redding LE, Morris DO, Mueller NT, Thorpe RJ Jr, Agnew J, Carroll KC, Planet PJ, Rubenstein RC, Chen AR, Grice EA, and Davis MF
- Abstract
Microbial sharing between humans and animals has been demonstrated in a variety of settings. However, the extent of microbial sharing that occurs within the healthcare setting during animal-assisted intervention programs is unknown. Understanding microbial transmission between patients and therapy dogs can provide important insights into potential health benefits for patients, in addition to addressing concerns regarding potential pathogen transmission that limits program utilization. This study evaluated for potential microbial sharing between pediatric patients and therapy dogs and tested whether patient-dog contact level and a dog decolonization protocol modified this sharing. Patients, therapy dogs, and the hospital environment were sampled before and after every group therapy session and samples underwent 16S rRNA sequencing to characterize microbial communities. Both patients and dogs experienced changes in the relative abundance and overall diversity of their nasal microbiome, suggesting that the exchange of microorganisms had occurred. Increased contact was associated with greater sharing between patients and therapy dogs, as well as between patients. A topical chlorhexidine-based dog decolonization was associated with decreased microbial sharing between therapy dogs and patients but did not significantly affect sharing between patients. These data suggest that the therapy dog is both a potential source of and a vehicle for the transfer of microorganisms to patients but not necessarily the only source. The relative contribution of other potential sources (e.g., other patients, the hospital environment) should be further explored to determine their relative importance.
- Published
- 2021
- Full Text
- View/download PDF
31. Bacteria induce skin regeneration via IL-1β signaling.
- Author
-
Wang G, Sweren E, Liu H, Wier E, Alphonse MP, Chen R, Islam N, Li A, Xue Y, Chen J, Park S, Chen Y, Lee S, Wang Y, Wang S, Archer NK, Andrews W, Kane MA, Dare E, Reddy SK, Hu Z, Grice EA, Miller LS, and Garza LA
- Subjects
- Adolescent, Adult, Animals, Bacteria classification, Bacteria genetics, Bacteria isolation & purification, Female, Humans, Interleukin-1beta genetics, Keratinocytes metabolism, Keratinocytes microbiology, Male, Mice, Mice, Inbred C57BL, Microbiota, Middle Aged, Myeloid Differentiation Factor 88 genetics, Myeloid Differentiation Factor 88 metabolism, Receptors, Interleukin-1 genetics, Receptors, Interleukin-1 metabolism, Regeneration, Signal Transduction, Skin metabolism, Wound Healing, Wounds and Injuries genetics, Wounds and Injuries metabolism, Young Adult, Interleukin-1beta metabolism, Skin microbiology, Skin physiopathology, Wounds and Injuries microbiology, Wounds and Injuries physiopathology
- Abstract
Environmental factors that enhance regeneration are largely unknown. The immune system and microbiome are attributed roles in repairing and regenerating structure but their precise interplay is unclear. Here, we assessed the function of skin bacteria in wound healing and wound-induced hair follicle neogenesis (WIHN), a rare adult organogenesis model. WIHN levels and stem cell markers correlate with bacterial counts, being lowest in germ-free (GF), intermediate in conventional specific pathogen-free (SPF), and highest in wild-type mice, even those infected with pathogenic Staphylococcus aureus. Reducing skin microbiota via cage changes or topical antibiotics decreased WIHN. Inflammatory cytokine IL-1β and keratinocyte-dependent IL-1R-MyD88 signaling are necessary and sufficient for bacteria to promote regeneration. Finally, in a small trial, a topical broad-spectrum antibiotic also slowed skin wound healing in adult volunteers. These results demonstrate a role for IL-1β to control morphogenesis and support the need to reconsider routine applications of topical prophylactic antibiotics., Competing Interests: Declaration of interests L.S.M. is a full-time employee of Janssen Pharmaceuticals and may hold Johnson & Johnson stock and stock options. L.S.M. performed all work at his prior affiliation at Johns Hopkins University School of Medicine, and he has received prior grant support from Astra Zeneca, Pfizer, Boehringer Ingelheim, Regeneron Pharmaceuticals, and Moderna Therapeutics; he was also a paid consultant for Armirall and Janssen Research and Development, was on the scientific advisory board of Integrated Biotherapeutics, and is a shareholder of Noveome Biotherapeutics, which are all developing therapeutics against infections (including S. aureus and other pathogens) and/or inflammatory conditions., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
32. Whole-Exome and Transcriptome Analysis of UV-Exposed Epidermis and Carcinoma In Situ Reveals Early Drivers of Carcinogenesis.
- Author
-
Zheng Q, Capell BC, Parekh V, O'Day C, Atillasoy C, Bashir HM, Yeh C, Shim EH, Prouty SM, Dentchev T, Lee V, Wushanley L, Kweon Y, Suzuki-Horiuchi Y, Pear W, Grice EA, and Seykora JT
- Subjects
- Carcinogenesis genetics, Carcinoma in Situ genetics, Carcinoma, Squamous Cell genetics, Genes, p53, Humans, Mutation, Neoplasms, Radiation-Induced genetics, Receptors, Notch genetics, Sequence Analysis, RNA, Skin Neoplasms genetics, Ultraviolet Rays, Carcinoma in Situ etiology, Carcinoma, Squamous Cell etiology, Epidermis radiation effects, Exome, Gene Expression Profiling, Neoplasms, Radiation-Induced etiology, Skin Neoplasms etiology
- Abstract
Squamous cell carcinoma in situ (SCCIS) is a prevalent precancerous lesion that can progress to cutaneous squamous cell carcinoma. Although SCCIS is common, its pathogenesis remains poorly understood. To better understand SCCIS development, we performed laser captured microdissection of human SCCIS and the adjacent epidermis to isolate genomic DNA and RNA for next-generation sequencing. Whole-exome sequencing identified UV-signature mutations in multiple genes, including NOTCH1-3 in the epidermis and SCCIS and oncogenic TP53 mutations in SCCIS. Gene families, including SLFN genes, contained UV/oxidative-signature disruptive epidermal mutations that manifested positive selection in SCCIS. The frequency and distribution of NOTCH and TP53 mutations indicate that NOTCH mutations may precede TP53 mutations. RNA sequencing identified 1,166 differentially expressed genes; the top five enriched gene ontology biological processes included (i) immune response, (ii) epidermal development, (iii) protein phosphorylation, (iv) regulation of catalytic activity, and (v) cytoskeletal regulation. The NEURL1 ubiquitin ligase, which targets Notch ligands for degradation, was upregulated in SCCIS. NEURL1 protein was found to be elevated in SCCIS suggesting that increased levels could represent a mechanism for downregulating Notch during UV-induced carcinogenesis. The data from DNA and RNA sequencing of epidermis and SCCIS provide insights regarding SCCIS formation., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
33. The Skin Microbiota: Balancing Risk and Reward.
- Author
-
Flowers L and Grice EA
- Subjects
- Bacteria classification, Ecosystem, Fungi classification, Host Microbial Interactions immunology, Host Microbial Interactions physiology, Humans, Symbiosis physiology, Viruses classification, Bacteria metabolism, Microbiota physiology, Skin microbiology, Skin Diseases microbiology
- Abstract
The skin microbiome is an ecosystem comprised of a multitude of microbial species interacting with their surroundings, including other microbes and host epithelial and immune cells. These interactions are the basis of important roles within the skin microbiome that provide benefit to the host, boosting multiple aspects of barrier function, a critical function of this essential organ. However, with reward always comes risk; resident skin microbes function in a context-dependent manner, set on the backdrop of a dynamic host and microbial milieu. Here, we discuss the reward of hosting a microbial ecosystem on the skin, including protection from pathogens and tuning of the skin microenvironment. We also give consideration to how these skin residents, often termed "commensals" can cause disorder, damage, and promote skin disease., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
34. The otic microbiota and mycobiota in a referral population of dogs in eastern USA with otitis externa.
- Author
-
Bradley CW, Lee FF, Rankin SC, Kalan LR, Horwinski J, Morris DO, Grice EA, and Cain CL
- Subjects
- 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, Dog Diseases microbiology, Ear Canal microbiology, Microbiota, Mycobiome, Otitis Externa microbiology
- 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., (© 2020 ESVD and ACVD.)
- Published
- 2020
- Full Text
- View/download PDF
35. Human macrophage response to microbial supernatants from diabetic foot ulcers.
- Author
-
Deusenbery CB, Kalan L, Meisel JS, Gardner SE, Grice EA, and Spiller KL
- Subjects
- Adult, Aged, Bacterial Infections pathology, Biopsy, Needle, Cells, Cultured, Cohort Studies, Culture Media, Diabetic Foot physiopathology, Enzyme-Linked Immunosorbent Assay, Female, Humans, Male, Microbiota drug effects, Middle Aged, Sensitivity and Specificity, Wound Healing genetics, Anti-Infective Agents pharmacology, Bacterial Infections mortality, Diabetic Foot microbiology, Macrophages metabolism, Microbiota genetics, Wound Healing drug effects
- Abstract
Diabetic foot ulcers (DFUs) are a major clinical problem exacerbated by prolonged bacterial infection. Macrophages, the primary innate immune cells, are multifunctional cells that regulate diverse processes throughout multiple phases of wound healing. To better understand the influence of microbial species on macrophage behavior, we cultured primary human monocyte-derived macrophages from four donors for 24 hours in media conditioned by bacteria and fungi (Pseudomonas aeruginosa, Corynebacterium amycolatum, Corynebacterium striatum, Staphylococcus aureus, Staphylococcus simulans, and Candida albicans) isolated from the DFUs of six patients. The effects of these microbe-derived signals on macrophage behavior were assessed by measuring the gene expression of a panel of 25 genes related to macrophage phenotype, angiogenesis, bacterial recognition, and cell survival, as well as secretion of two inflammatory cytokines using NanoString multiplex analysis. Principal component analysis showed that macrophage gene expression and protein secretion were affected by both microbial species as well as human donor. S. simulans and C. albicans caused up-regulation of genes associated with a proinflammatory (M1) phenotype, and P. aeruginosa caused an increase in the secretion of the proinflammatory cytokine and M1 marker tumor necrosis factor-alpha (TNFα). Together, these results suggest that macrophages respond to secreted factors from microbes by up-regulating inflammatory markers, and that the effects are strongly dependent on the monocyte donor. Ultimately, increased understanding of macrophage-microbe interactions will lead to the development of more targeted therapies for DFU healing., (© 2019 by the Wound Healing Society.)
- Published
- 2019
- Full Text
- View/download PDF
36. Expanding the scope and scale of microbiome research.
- Author
-
Knight R, Ley RE, Raes J, and Grice EA
- Subjects
- High-Throughput Nucleotide Sequencing, Humans, Sequence Analysis, DNA, Microbiota genetics
- Published
- 2019
- Full Text
- View/download PDF
37. Strain- and Species-Level Variation in the Microbiome of Diabetic Wounds Is Associated with Clinical Outcomes and Therapeutic Efficacy.
- Author
-
Kalan LR, Meisel JS, Loesche MA, Horwinski J, Soaita I, Chen X, Uberoi A, Gardner SE, and Grice EA
- Subjects
- Animals, Coinfection therapy, Diabetic Foot therapy, Disease Models, Animal, Longitudinal Studies, Mice, Prospective Studies, Treatment Outcome, Wound Healing, Wound Infection therapy, Anti-Infective Agents therapeutic use, Coinfection microbiology, Debridement, Diabetic Foot microbiology, Microbiota, Wound Infection microbiology
- 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., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
38. Research Techniques Made Simple: Profiling the Skin Microbiota.
- Author
-
Grogan MD, Bartow-McKenney C, Flowers L, Knight SAB, Uberoi A, and Grice EA
- Subjects
- Bacteria isolation & purification, Dermatitis microbiology, Dermatitis pathology, Humans, Sequence Analysis, DNA, Skin pathology, Bacteria genetics, Biomedical Research methods, Dermatitis genetics, High-Throughput Nucleotide Sequencing methods, Metagenome genetics, Microbiota genetics, Skin microbiology
- Abstract
Skin is colonized by microbial communities (microbiota) that participate in immune homeostasis, development and maintenance of barrier function, and protection from pathogens. The past decade has been marked by an increased interest in the skin microbiota and its role in cutaneous health and disease, in part due to advances in next-generation sequencing platforms that enable high-throughput, culture-independent detection of bacteria, fungi, and viruses. Various approaches, including bacterial 16S ribosomal RNA gene sequencing and metagenomic shotgun sequencing, have been applied to profile microbial communities colonizing healthy skin and diseased skin including atopic dermatitis, psoriasis, and acne, among others. Here, we provide an overview of culture-dependent and -independent approaches to profiling the skin microbiota and the types of questions that may be answered by each approach. We additionally highlight important study design considerations, selection of controls, interpretation of results, and limitations and challenges., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
39. Antiseptic Agents Elicit Short-Term, Personalized, and Body Site-Specific Shifts in Resident Skin Bacterial Communities.
- Author
-
SanMiguel AJ, Meisel JS, Horwinski J, Zheng Q, Bradley CW, and Grice EA
- Subjects
- Administration, Cutaneous, Adult, Bacteria isolation & purification, Female, Healthy Volunteers, Humans, Male, Polymerase Chain Reaction, Skin drug effects, Skin Diseases, Bacterial microbiology, Young Adult, Anti-Infective Agents, Local administration & dosage, Bacteria genetics, DNA, Bacterial analysis, Microbiota drug effects, Skin microbiology, Skin Diseases, Bacterial prevention & control
- 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., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
40. Longitudinal Study of the Psoriasis-Associated Skin Microbiome during Therapy with Ustekinumab in a Randomized Phase 3b Clinical Trial.
- Author
-
Loesche MA, Farahi K, Capone K, Fakharzadeh S, Blauvelt A, Duffin KC, DePrimo SE, Muñoz-Elías EJ, Brodmerkel C, Dasgupta B, Chevrier M, Smith K, Horwinski J, Tyldsley A, and Grice EA
- Subjects
- 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, Bacteria genetics, Microbiota drug effects, Psoriasis drug therapy, Skin microbiology, Ustekinumab administration & dosage
- 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., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
41. Fungi in the Wound Microbiome.
- Author
-
Kalan L and Grice EA
- Abstract
Significance: Culture-independent methods have revealed the diverse and dynamic bacterial communities that colonize chronic wounds. Only recently have studies begun to examine fungal colonization and interactions with the bacterial component of the microbiome, their relationship with the host, and influence on wound outcomes. Recent Advances: Studies using culture-independent sequencing methods reveal that fungi often go undetected in wounds. Candida spp. and Cladosporidium spp. are the most commonly identified fungi in wounds. The wound environment may promote multispecies biofilm formation between bacteria and fungi in wounds, with implications for pathogenicity, treatment, and outcomes. Critical Issues: Identifying microorganisms that are problematic for healing will require a comprehensive understanding of all members of the polymicrobial wound community, including fungi and bacteria. Improved reference databases and bioinformatics tools for studying fungal communities will stimulate further research into the fungal microbiome. Future Directions: Continued study of polymicrobial wound communities using culture-independent methods will further our understanding of the relationships between microbial bioburden, the host response, and impact on healing, complications, and patient outcomes. Future studies should encompass all types of microbiota, including fungi, and focus on potential multi-kingdom interactions that contribute to pathogenicity, biofilm formation, and poor outcomes.
- Published
- 2018
- Full Text
- View/download PDF
42. HmmUFOtu: An HMM and phylogenetic placement based ultra-fast taxonomic assignment and OTU picking tool for microbiome amplicon sequencing studies.
- Author
-
Zheng Q, Bartow-McKenney C, Meisel JS, and Grice EA
- Subjects
- Algorithms, Cluster Analysis, Computational Biology, High-Throughput Nucleotide Sequencing methods, Phylogeny, RNA, Ribosomal, 16S genetics, Sequence Analysis, DNA methods, Microbiota genetics
- Abstract
Culture-independent analysis of microbial communities frequently relies on amplification and sequencing of the prokaryotic 16S ribosomal RNA gene. Typical analysis pipelines group sequences into operational taxonomic units (OTUs) to infer taxonomic and phylogenetic relationships. Here, we present HmmUFOtu, a novel tool for processing microbiome amplicon sequencing data, which performs rapid per-read phylogenetic placement, followed by phylogenetically informed clustering into OTUs and taxonomy assignment. Compared to standard pipelines, HmmUFOtu more accurately and reliably recapitulates microbial community diversity and composition in simulated and real datasets without relying on heuristics or sacrificing speed or accuracy.
- Published
- 2018
- Full Text
- View/download PDF
43. Severe pain during wound care procedures: A cross-sectional study protocol.
- Author
-
Fiala CA, Abbott LI, Carter CD, Hillis SL, Wolf JS, Schuster M, Dulski R, Grice EA, Rakel BA, and Gardner SE
- Abstract
Aim: The aim of this study is to: (a) develop and evaluate a model to predict severe pain during wound care procedures (WCPs) so that high-risk patients can be targeted for specialized dressings and preventive pain control; and (b) identify biological factors associated with severe pain during WCPs so that novel pain control strategies can be developed., Background: Wound care procedures such as dressing changes can cause moderate to severe pain in 74% of patients, with nearly half (36%) of all patients experiencing severe pain (rated as 8-10 on a 10-point numeric rating scale) during dressing change. Additionally, clinicians have little direction with current guidelines regarding pain control during WCPs including the selection of the appropriate advanced wound dressings and the appropriate use of analgesics., Design: This is a cross-sectional study., Methods: The National Institute of Nursing Research approved and funded the study June of 2015 and the appropriate Institutional Review Board approved all study protocols prior to funding. Study enrolment is underway at the University of Iowa Hospitals and Clinics with a target of 525 participants. Potential participants must be adults (21+ years) and have a nonburn, nondiabetic foot, full-thickness wound. The research team performs a one-time study dressing change on enrolled participants and collects all study data., Discussion: This study will allow the development of a tool for clinicians to use to predict severe pain during WCPs and identify biological factors significantly associated with severe pain during WCPs., (© 2018 John Wiley & Sons Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
44. The microbiota of traumatic, open fracture wounds is associated with mechanism of injury.
- Author
-
Bartow-McKenney C, Hannigan GD, Horwinski J, Hesketh P, Horan AD, Mehta S, and Grice EA
- Subjects
- 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, Bacteria classification, Fractures, Open microbiology, Microbiota physiology, Skin microbiology, Wound Healing physiology, Wound Infection microbiology
- 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., (© 2018 by the Wound Healing Society.)
- Published
- 2018
- Full Text
- View/download PDF
45. Commensal microbiota modulate gene expression in the skin.
- Author
-
Meisel JS, Sfyroera G, Bartow-McKenney C, Gimblet C, Bugayev J, Horwinski J, Kim B, Brestoff JR, Tyldsley AS, Zheng Q, Hodkinson BP, Artis D, and Grice EA
- Subjects
- 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, Gastrointestinal Tract microbiology, Gene Expression Profiling methods, Gene Regulatory Networks, Skin microbiology
- 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.
- Published
- 2018
- Full Text
- View/download PDF
46. Host-microbe interactions: Malassezia and human skin.
- Author
-
Grice EA and Dawson TL 23⁎Jr
- Subjects
- Animals, Dermatomycoses physiopathology, Host-Pathogen Interactions, Humans, Malassezia genetics, Skin microbiology, Skin Diseases, Bacterial physiopathology, Dermatomycoses microbiology, Malassezia physiology, Skin Diseases, Bacterial microbiology
- Abstract
The skin is our first line of defense, protecting us from invasion and evaporation. Its variable structure, changing geography, and complex immune repertoire provide a vast interface for our cutaneous microbial community. Skin is inhabited by many thousands of microbes, but this review focuses on the dominant eukaryote, Malassezia, and its host interaction. Malassezia compromises 17 species with variable niche specificities and differing pathogenic potential. It has been known as a skin inhabitant for over 100 years, and is now accepted to be on all warm-blooded animals. Malassezia occupy healthy and diseased skin, so their role as commensal or pathogenic organisms is complex. Malassezia interact with their host indirectly through immune interplay and directly via chemical mediators. While some interactions are known, many remain to be fully understood., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
47. The preadolescent acne microbiome: A prospective, randomized, pilot study investigating characterization and effects of acne therapy.
- Author
-
Coughlin CC, Swink SM, Horwinski J, Sfyroera G, Bugayev J, Grice EA, and Yan AC
- Subjects
- Acne Vulgaris microbiology, Administration, Topical, Child, Female, Humans, Male, Microbiota genetics, Phylogeny, Pilot Projects, Prospective Studies, Skin microbiology, Treatment Outcome, Acne Vulgaris drug therapy, Benzoyl Peroxide administration & dosage, Dermatologic Agents administration & dosage, Keratolytic Agents administration & dosage, Microbiota drug effects, Tretinoin administration & dosage
- 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., (© 2017 Wiley Periodicals, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
48. Cutaneous Burn Injury Promotes Shifts in the Bacterial Microbiome in Autologous Donor Skin: Implications for Skin Grafting Outcomes.
- Author
-
Plichta JK, Gao X, Lin H, Dong Q, Toh E, Nelson DE, Gamelli RL, Grice EA, and Radek KA
- Subjects
- Adult, Autografts, Female, Humans, Male, Middle Aged, Bacterial Infections etiology, Bacterial Infections microbiology, Bacterial Infections mortality, Bacterial Infections therapy, Burns microbiology, Burns mortality, Burns surgery, Gastrointestinal Microbiome, Graft Survival, Sepsis etiology, Sepsis microbiology, Sepsis mortality, Sepsis therapy, Skin Transplantation, Wound Infection microbiology, Wound Infection mortality, Wound Infection therapy
- Abstract
Introduction: The cutaneous microbiome maintains skin barrier function, regulates inflammation, and stimulates wound-healing responses. Burn injury promotes an excessive activation of the cutaneous and systemic immune response directed against commensal and invading pathogens. Skin grafting is the primary method of reconstructing full-thickness burns, and wound infection continues to be a significant complication., Methods: In this study, the cutaneous bacterial microbiome was evaluated and subsequently compared to patient outcomes. Three different full-thickness skin specimens were assessed: control skin from non-burned subjects; burn margin from burn patients; and autologous donor skin from the same cohort of burn patients., Results: We observed that skin bacterial community structure of burn patients was significantly altered compared with control patients. We determined that the unburned autologous donor skin from burn patients exhibits a microbiome similar to that of the burn margin, rather than unburned controls, and that changes in the cutaneous microbiome statistically correlate with several post-burn complications. We established that Corynebacterium positively correlated with burn wound infection, while Staphylococcus and Propionibacterium negatively correlated with burn wound infection. Both Corynebacterium and Enterococcus negatively correlated with the development of sepsis., Conclusions: This study identifies distinct differences in the cutaneous microbiome between burn subjects and unburned controls, and ascertains that select bacterial taxa significantly correlate with several comorbid complications of burn injury. These preliminary data suggest that grafting donor skin exhibiting bacterial dysbiosis may augment infection and/or graft failure and sets the foundation for more in-depth and mechanistic analyses in presumably "healthy" donor skin from patients requiring skin grafting procedures.
- Published
- 2017
- Full Text
- View/download PDF
49. Topical Antimicrobial Treatments Can Elicit Shifts to Resident Skin Bacterial Communities and Reduce Colonization by Staphylococcus aureus Competitors.
- Author
-
SanMiguel AJ, Meisel JS, Horwinski J, Zheng Q, and Grice EA
- Subjects
- Administration, Cutaneous, Animals, Anti-Infective Agents, Local pharmacology, Female, Mice, Microbiota drug effects, Anti-Bacterial Agents pharmacology, Skin microbiology, Staphylococcal Skin Infections drug therapy, Staphylococcus aureus drug effects
- 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., (Copyright © 2017 American Society for Microbiology.)
- Published
- 2017
- Full Text
- View/download PDF
50. Cutaneous Leishmaniasis Induces a Transmissible Dysbiotic Skin Microbiota that Promotes Skin Inflammation.
- Author
-
Gimblet C, Meisel JS, Loesche MA, Cole SD, Horwinski J, Novais FO, Misic AM, Bradley CW, Beiting DP, Rankin SC, Carvalho LP, Carvalho EM, Scott P, and Grice EA
- Subjects
- Animals, Disease Models, Animal, Humans, Hypersensitivity, Leishmania major immunology, Leishmania major pathogenicity, Mice, Mice, Inbred C57BL, Microbiota immunology, Skin microbiology, Skin parasitology, Staphylococcus immunology, Staphylococcus pathogenicity, Streptococcus immunology, Streptococcus pathogenicity, Dysbiosis etiology, Dysbiosis immunology, Inflammation immunology, Inflammation microbiology, Leishmania braziliensis pathogenicity, Leishmaniasis, Cutaneous complications, Leishmaniasis, Cutaneous microbiology, Microbiota physiology, Skin immunology
- 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., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.