Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Host Microbe ; 32(8): 1264-1279, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39146798

RESUMEN

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.


Asunto(s)
Microbiota , Piel , Simbiosis , Humanos , Piel/microbiología , Microbiota/fisiología , Enfermedades de la Piel/microbiología , Animales , Homeostasis , Interacciones Microbiota-Huesped , Bacterias/metabolismo
2.
Sci Adv ; 10(26): eadj2020, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38924411

RESUMEN

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.


Asunto(s)
Alcaligenes faecalis , Queratinocitos , Metaloproteinasas de la Matriz , Cicatrización de Heridas , Alcaligenes faecalis/metabolismo , Animales , Queratinocitos/metabolismo , Queratinocitos/microbiología , Humanos , Metaloproteinasas de la Matriz/metabolismo , Metaloproteinasas de la Matriz/genética , Pie Diabético/microbiología , Pie Diabético/patología , Pie Diabético/metabolismo , Ratones , Repitelización , Masculino
3.
Vet Dermatol ; 35(4): 375-385, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38616572

RESUMEN

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.


Asunto(s)
Ciclosporina , Enfermedades de los Perros , Cetoconazol , Fístula Rectal , Animales , Perros , Ciclosporina/uso terapéutico , Ciclosporina/administración & dosificación , Enfermedades de los Perros/tratamiento farmacológico , Enfermedades de los Perros/microbiología , Masculino , Cetoconazol/uso terapéutico , Cetoconazol/administración & dosificación , Femenino , Fístula Rectal/veterinaria , Fístula Rectal/tratamiento farmacológico , Fístula Rectal/microbiología , Estudios Longitudinales , Recto/microbiología , Piel/microbiología , Piel/patología , Microbiota/efectos de los fármacos
4.
Nat Rev Microbiol ; 22(8): 507-521, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38575708

RESUMEN

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.


Asunto(s)
Microbiota , Cicatrización de Heridas , Infección de Heridas , Humanos , Infección de Heridas/microbiología , Piel/microbiología , Biopelículas/crecimiento & desarrollo , Animales
5.
Cell Rep ; 43(4): 114029, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38573852

RESUMEN

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.


Asunto(s)
Microbiota , Fenotipo , Linfocitos T , Animales , Ratones , Linfocitos T/inmunología , Linfocitos T/metabolismo , Masculino , Femenino , Ratones Endogámicos C57BL
6.
mSphere ; 9(3): e0063623, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38415632

RESUMEN

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.


Asunto(s)
Antiinfecciosos , Carnobacteriaceae , Staphylococcus aureus Resistente a Meticilina , Humanos , Porcinos , Animales , Ratones , Staphylococcus aureus , Proteómica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA