Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 55(9): 1645-1662.e7, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-35882236

RESUMEN

Healthy skin maintains a diverse microbiome and a potent immune system to fight off infections. Here, we discovered that the epithelial-cell-derived antimicrobial peptides defensins activated orphan G-protein-coupled receptors (GPCRs) Mrgpra2a/b on neutrophils. This signaling axis was required for effective neutrophil-mediated skin immunity and microbiome homeostasis. We generated mutant mouse lines lacking the entire Defensin (Def) gene cluster in keratinocytes or Mrgpra2a/b. Def and Mrgpra2 mutant animals both exhibited skin dysbiosis, with reduced microbial diversity and expansion of Staphylococcus species. Defensins and Mrgpra2 were critical for combating S. aureus infections and the formation of neutrophil abscesses, a hallmark of antibacterial immunity. Activation of Mrgpra2 by defensin triggered neutrophil release of IL-1ß and CXCL2 which are vital for proper amplification and propagation of the antibacterial immune response. This study demonstrated the importance of epithelial-neutrophil signaling via the defensin-Mrgpra2 axis in maintaining healthy skin ecology and promoting antibacterial host defense.


Asunto(s)
Infecciones Bacterianas , Neutrófilos , Receptores Acoplados a Proteínas G , Animales , Ratones , Antibacterianos , Proteínas Portadoras , Defensinas/genética , Disbiosis , Queratinocitos , Receptores Acoplados a Proteínas G/metabolismo , Staphylococcus aureus
2.
Exp Dermatol ; 32(4): 425-435, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36461082

RESUMEN

Phosphodiesterase 4 (PDE4) is highly expressed in keratinocytes and immune cells and promotes pro-inflammatory responses upon activation. The activity of PDE4 has been attributed to various inflammatory conditions, leading to the development and approval of PDE4 inhibitors as host-directed therapeutics in humans. For example, the topical PDE4 inhibitor, crisaborole, is approved for the treatment of mild-to-moderate atopic dermatitis and has shown efficacy in patients with psoriasis. However, the role of crisaborole in regulating the immunopathogenesis of inflammatory skin diseases and infection is not entirely known. Therefore, we evaluated the effects of crisaborole in multiple mouse models, including psoriasis-like dermatitis, AD-like skin inflammation with and without filaggrin mutations, and Staphylococcus aureus skin infection. We discovered that crisaborole dampens myeloid cells and itch in the skin during psoriasis-like dermatitis. Furthermore, crisaborole was effective in reducing skin inflammation in the context of filaggrin deficiency. Importantly, crisaborole reduced S. aureus skin colonization during AD-like skin inflammation. However, crisaborole was not efficacious in treating S. aureus skin infections, even as adjunctive therapy to antibiotics. Taken together, we found that crisaborole reduced itch during psoriasis-like dermatitis and decreased S. aureus skin colonization upon AD-like skin inflammation, which act as additional mechanisms by which crisaborole dampens the immunopathogenesis in mouse models of inflammatory skin diseases. Further examination is warranted to translate these preclinical findings to human disease.


Asunto(s)
Dermatitis Atópica , Inhibidores de Fosfodiesterasa 4 , Psoriasis , Infecciones Estafilocócicas , Humanos , Animales , Ratones , Staphylococcus aureus , Proteínas Filagrina , Modelos Animales de Enfermedad , Dermatitis Atópica/tratamiento farmacológico , Inhibidores de Fosfodiesterasa 4/uso terapéutico , Prurito/tratamiento farmacológico , Psoriasis/tratamiento farmacológico , Infecciones Estafilocócicas/tratamiento farmacológico , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4 , Inflamación/tratamiento farmacológico
3.
Exp Dermatol ; 32(7): 1051-1062, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37039485

RESUMEN

Cyclic AMP (cAMP) has a key role in psoriasis pathogenesis, as indicated by the therapeutic efficacy of phosphodiesterase inhibitors that prevent the degradation of cAMP. However, whether soluble adenylate cyclase (sAC) (encoded by the ADCY10 gene), which is an important source for cAMP, is involved in Th17 cell-mediated inflammation or could be an alternative therapeutic target in psoriasis is unknown. We have utilized the imiquimod model of murine psoriasiform dermatitis to address this question. Adcy10-/- mice had reduced erythema, scaling and swelling in the skin and reduced CD4+ IL17+ cell numbers in the draining lymph nodes, compared with wild-type mice after induction of psoriasiform dermatitis with imiquimod. Keratinocyte-specific knock out of Adcy10 had no effect on imiquimod-induced ear swelling suggesting keratinocyte sAC has no role in imiquimod-induced inflammation. During Th17 polarization in vitro, naive T cells from Adcy10-/- mice exhibited reduced IL17 secretion and IL-17+ T-cell proliferation suggesting that differentiation into Th17 cells is suppressed without sAC activity. Interestingly, loss of sAC did not impact the expression of Th17 lineage-defining transcription factors (such as Rorc and cMaf) but rather was required for CREB-dependent gene expression, which is known to support Th17 cell gene expression. Finally, topical application of small molecule sAC inhibitors (sACi) reduced imiquimod-induced psoriasiform dermatitis and Il17 gene expression in the skin. Collectively, these findings demonstrate that sAC is important for psoriasiform dermatitis in mouse skin. sACi may provide an alternative class of topical therapeutics for Th17-mediated skin diseases.


Asunto(s)
Adenilil Ciclasas , Eccema , Psoriasis , Animales , Ratones , Adenilil Ciclasas/genética , Adenilil Ciclasas/metabolismo , Modelos Animales de Enfermedad , Eccema/patología , Imiquimod/efectos adversos , Inflamación/tratamiento farmacológico , Inflamación/patología , Psoriasis/inducido químicamente , Psoriasis/tratamiento farmacológico , Psoriasis/metabolismo , Piel/metabolismo , Células Th17/metabolismo
4.
Drug Dev Res ; 84(8): 1567-1571, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37540034

RESUMEN

Staphylococcus aureus is the leading cause of skin and soft tissue infections. With the emergence of antibiotic-resistant bacteria, there is an unmet clinical need to develop immune-based therapies to treat skin infections. Previously, we have shown pan-caspase inhibition as a potential host-directed immunotherapy against community-acquired methicillin-resistant S aureus (CA-MRSA) and other bacterial skin infections. Here, we evaluated the role of irreversible pan-caspase inhibitor emricasan as a monotherapy and an adjunctive with a standard-of-care antibiotic, doxycycline, as potential host-directed immunotherapies against S. aureus skin infections in vivo. We used the established CA-MRSA strain USA300 on the dorsum of WT C57BL/6J mice and monitored lesion size and bacterial burden noninvasively, and longitudinally over 14 days with in vivo bioluminescence imaging (BLI). Mice in four groups placebo (0.5% carboxymethyl cellulose [CMC] solution), placebo plus doxycycline (100 mg/kg), emricasan (40 mg/kg) plus doxycycline, and emricasan only were treated orally twice daily by oral gavage for 7 days, starting at 4 h after injection of S aureus. When compared with placebo, all three groups, placebo plus doxycycline, emricasan plus doxycycline, and emricasan treated group, exhibited biological effect, with reduction of both the lesion size (*p = .0277, ****p < .0001, ****p < .0001, respectively) and bacterial burden (***p = .003, ****p < .0001, ****p < .0001, respectively). Importantly, the efficacy of emricasan against S. aureus was not due to direct antibacterial activity. Collectively, pan-caspase inhibitor emricasan and emricasan plus doxycycline reduced both the lesion size and bacterial burden in vivo, and emricasan is a potential host-directed immunotherapy against MRSA skin infections in a preclinical mouse model.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Infecciones Cutáneas Estafilocócicas , Ratones , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Doxiciclina/farmacología , Doxiciclina/uso terapéutico , Staphylococcus aureus , Infecciones Cutáneas Estafilocócicas/tratamiento farmacológico , Infecciones Cutáneas Estafilocócicas/microbiología , Ratones Endogámicos C57BL
5.
Proc Natl Acad Sci U S A ; 116(22): 10917-10926, 2019 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-31088972

RESUMEN

T cell cytokines contribute to immunity against Staphylococcus aureus, but the predominant T cell subsets involved are unclear. In an S. aureus skin infection mouse model, we found that the IL-17 response was mediated by γδ T cells, which trafficked from lymph nodes to the infected skin to induce neutrophil recruitment, proinflammatory cytokines IL-1α, IL-1ß, and TNF, and host defense peptides. RNA-seq for TRG and TRD sequences in lymph nodes and skin revealed a single clonotypic expansion of the encoded complementarity-determining region 3 amino acid sequence, which could be generated by canonical nucleotide sequences of TRGV5 or TRGV6 and TRDV4 However, only TRGV6 and TRDV4 but not TRGV5 sequences expanded. Finally, Vγ6+ T cells were a predominant γδ T cell subset that produced IL-17A as well as IL-22, TNF, and IFNγ, indicating a broad and substantial role for clonal Vγ6+Vδ4+ T cells in immunity against S. aureus skin infections.


Asunto(s)
Interleucina-17/fisiología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/patogenicidad , Subgrupos de Linfocitos T/inmunología , Linfocitos T/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Ganglios Linfáticos/inmunología , Ratones , Infecciones Estafilocócicas/microbiología
6.
J Cell Mol Med ; 25(21): 10008-10019, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34623736

RESUMEN

Fibrosis is a major health burden across diseases and organs. To remedy this, we study wound-induced hair follicle neogenesis (WIHN) as a model of non-fibrotic healing that recapitulates embryogenesis for de novo hair follicle morphogenesis after wounding. We previously demonstrated that TLR3 promotes WIHN through binding wound-associated dsRNA, the source of which is still unclear. Here, we find that multiple distinct contexts of high WIHN all show a strong neutrophil signature. Given the correlation between neutrophil infiltration and endogenous dsRNA release, we hypothesized that neutrophil extracellular traps (NETs) likely release nuclear spliceosomal U1 dsRNA and modulate WIHN. However, rather than enhance regeneration, we find mature neutrophils inhibit WIHN such that mice with mature neutrophil depletion exhibit higher WIHN. Similarly, Pad4 null mice, which are defective in NET production, show augmented WIHN. Finally, using single-cell RNA sequencing, we identify a dramatic increase in mature and activated neutrophils in the wound beds of low regenerating Tlr3-/- mice. Taken together, these results demonstrate that although mature neutrophils are stimulated by a common pro-regenerative cue, their presence and NETs hinder regeneration.


Asunto(s)
Trampas Extracelulares , Neutrófilos/inmunología , Neutrófilos/metabolismo , Regeneración , Animales , Biomarcadores , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Humanos , Inmunofenotipificación , Ratones , Ratones Noqueados , Infiltración Neutrófila , Análisis de la Célula Individual/métodos , Piel/metabolismo , Cicatrización de Heridas/genética , Cicatrización de Heridas/inmunología
7.
Emerg Infect Dis ; 27(3): 740-748, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33622471

RESUMEN

Transmission of livestock-associated Staphylococcus aureus clonal complex 9 (LA-SA CC9) between pigs raised on industrial hog operations (IHOs) and humans in the United States is poorly understood. We analyzed whole-genome sequences from 32 international S. aureus CC9 isolates and 49 LA-SA CC9 isolates from IHO pigs and humans who work on or live near IHOs in 10 pig-producing counties in North Carolina, USA. Bioinformatic analysis of sequence data from the 81 isolates demonstrated 3 major LA-SA CC9 clades. North Carolina isolates all fell within a single clade (C3). High-resolution phylogenetic analysis of C3 revealed 2 subclades of intermingled IHO pig and human isolates differing by 0-34 single-nucleotide polymorphisms. Our findings suggest that LA-SA CC9 from pigs and humans share a common source and provide evidence of transmission of antimicrobial-resistant LA-SA CC9 between IHO pigs and humans who work on or live near IHOs in North Carolina.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Crianza de Animales Domésticos , Animales , Antibacterianos , Humanos , Ganado , North Carolina , Filogenia , Staphylococcus aureus , Porcinos , Estados Unidos
8.
Immunity ; 37(4): 747-58, 2012 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-23063331

RESUMEN

Scratching triggers skin flares in atopic dermatitis. We demonstrate that scratching of human skin and tape stripping of mouse skin cause neutrophil influx. In mice, this influx was largely dependent on the generation of leukotriene B4 (LTB4) by neutrophils and their expression of the LTB4 receptor BLT1. Allergic skin inflammation in response to epicutaneous (EC) application of ovalbumin to tape-stripped skin was severely impaired in Ltb4r1(-/-) mice and required expression of BLT1 on both T cells and non-T cells. Cotransfer of wild-type (WT) neutrophils, but not neutrophils deficient in BLT1 or the LTB4-synthesizing enzyme LTA4H, restored the ability of WT CD4(+) effector T cells to transfer allergic skin inflammation to Ltb4r1(-/-) recipients. Pharmacologic blockade of LTB4 synthesis inhibited allergic skin inflammation elicited by cutaneous antigen challenge in previously EC-sensitized mice. Our results demonstrate that a neutrophil-T cell axis reliant on LTB4-BLT1 interaction is required for allergic skin inflammation.


Asunto(s)
Dermatitis/inmunología , Leucotrieno B4/inmunología , Infiltración Neutrófila , Neutrófilos/inmunología , Animales , Biopsia , Dermatitis/patología , Modelos Animales de Enfermedad , Humanos , Leucotrieno B4/biosíntesis , Ratones , Ratones Endogámicos C57BL , Receptores de Leucotrieno B4/deficiencia , Receptores de Leucotrieno B4/inmunología
9.
Proc Natl Acad Sci U S A ; 115(47): E11111-E11119, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30297395

RESUMEN

Staphylococcus aureus is the leading cause of skin and skin structure infection (SSSI), a primary portal of entry for invasive infection. Our prior studies discovered a role for protective innate memory against recurrent methicillin-resistant S. aureus (MRSA) SSSI. In the present study, the dynamics and mechanisms of this response were explored in recurrent SSSI in WT mice. Priming by prior infection reduced skin lesion severity and MRSA burden, and protected against dissemination at day 7 but not day 2. Cytokine and cellular signatures in SSSI differed at day 2 versus 7, and were distinct in skin versus blood or spleen. Cytokines associated with protection in skin included increased IL-17, IL-6, monokine inducible by IFN-γ (MIG), and RANTES, while increased IP-10 correlated with protection from dissemination. Cellular signatures of protection included increased Th17, M1 macrophage, and dendritic cell populations in abscesses, and total macrophages in lymph nodes. Priming potentiated S. aureus-specific phagocytic killing by bone marrow-derived macrophages in vitro, and their adoptive transfer into naïve skin afforded protective efficacy in vivo. Present findings indicate that protective immunity in recurrent S. aureus infection is locally targeted, and involves specific memory conferred by macrophages. These insights provide targets for vaccine and immunotherapeutic development against MRSA.


Asunto(s)
Inmunidad Innata/inmunología , Memoria Inmunológica/inmunología , Macrófagos/inmunología , Macrófagos/trasplante , Staphylococcus aureus Resistente a Meticilina/inmunología , Infecciones Cutáneas Estafilocócicas/inmunología , Traslado Adoptivo , Animales , Quimiocina CCL5/sangre , Quimiocina CXCL10/sangre , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Proteínas de Homeodominio/genética , Interferón gamma/sangre , Interleucina-17/sangre , Interleucina-6/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones Cutáneas Estafilocócicas/microbiología , Células Th17/inmunología
10.
EMBO Rep ; 19(12)2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30337494

RESUMEN

Chitin is the second most abundant polysaccharide in nature and linked to fungal infection and asthma. However, bona fide immune receptors directly binding chitin and signaling immune activation and inflammation have not been clearly identified because polymeric crude chitin with unknown purity and molecular composition has been used. By using defined chitin (N-acetyl-glucosamine) oligomers, we here identify six-subunit-long chitin chains as the smallest immunologically active motif and the innate immune receptor Toll-like receptor (TLR2) as a primary fungal chitin sensor on human and murine immune cells. Chitin oligomers directly bind TLR2 with nanomolar affinity, and this fungal TLR2 ligand shows overlapping and distinct signaling outcomes compared to known mycobacterial TLR2 ligands. Unexpectedly, chitin oligomers composed of five or less subunits are inactive, hinting to a size-dependent system of immuno-modulation that appears conserved in plants and humans. Since blocking of the chitin-TLR2 interaction effectively prevents chitin-mediated inflammation in vitro and in vivo, our study highlights the chitin-TLR2 interaction as a potential target for developing novel therapies in chitin-related pathologies and fungal disease.


Asunto(s)
Quitina/química , Quitina/metabolismo , Hongos/metabolismo , Inflamación/metabolismo , Inflamación/patología , Receptor Toll-Like 2/metabolismo , Animales , Pared Celular/efectos de los fármacos , Pared Celular/metabolismo , Quitinasas/metabolismo , Femenino , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Factores Inmunológicos/farmacología , Ligandos , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Células THP-1 , Receptor Toll-Like 1/agonistas , Receptor Toll-Like 1/metabolismo , Receptor Toll-Like 2/química , Zimosan/metabolismo
11.
J Immunol ; 201(6): 1651-1661, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30045969

RESUMEN

IL-17 is a potent proinflammatory cytokine that drives pathogenesis of multiple autoimmune diseases, including psoriasis. A major source of pathogenic IL-17 is a subset of γδ T cells (Tγδ17) that acquires the ability to produce IL-17 while developing in the thymus. The mechanisms that regulate homeostasis of Tγδ17 cells and their roles in psoriasis, however, are not fully understood. In this paper, we show that the heparan sulfate proteoglycan syndecan-1 (sdc1) plays a critical role in regulating homeostasis of Tγδ17 cells and modulating psoriasis-like skin inflammation in mice. sdc1 was predominantly expressed by Tγδ17 cells (but not IL-17- Tγδ cells) in the thymus, lymph nodes, and dermis. sdc1 deficiency significantly and selectively increased the frequency and absolute numbers of Tγδ17 cells by mechanisms that included increased proliferation and decreased apoptosis. Adoptive transfer experiments ruled out a significant role of sdc1 expressed on nonhematopoietic cells in halting expansion and proliferation of sdc1-deficient Tγδ17 cells. When subjected to imiquimod-induced psoriasiform dermatitis, Tγδ17 cells in sdc1KO mice displayed heightened responses accompanied by significantly increased skin inflammation than their wild-type counterparts. Furthermore, transferred sdc1-deficient γδ T cells caused more severe psoriasiform dermatitis than their sdc1-sufficient counterparts in TCR-ßδ KO hosts. The results uncover a novel role for sdc1 in controlling homeostasis of Tγδ17 cells and moderating host responses to psoriasis-like inflammation.


Asunto(s)
Dermatitis/inmunología , Interleucina-17/inmunología , Psoriasis/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Sindecano-1/inmunología , Linfocitos T/inmunología , Animales , Dermatitis/genética , Dermatitis/patología , Modelos Animales de Enfermedad , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-17/genética , Ratones , Ratones Noqueados , Psoriasis/genética , Psoriasis/patología , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Sindecano-1/genética , Linfocitos T/patología
12.
Proc Natl Acad Sci U S A ; 114(26): E5094-E5102, 2017 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-28607050

RESUMEN

Infection is a major complication of implantable medical devices, which provide a scaffold for biofilm formation, thereby reducing susceptibility to antibiotics and complicating treatment. Hematogenous implant-related infections following bacteremia are particularly problematic because they can occur at any time in a previously stable implant. Herein, we developed a model of hematogenous infection in which an orthopedic titanium implant was surgically placed in the legs of mice followed 3 wk later by an i.v. exposure to Staphylococcus aureus This procedure resulted in a marked propensity for a hematogenous implant-related infection comprised of septic arthritis, osteomyelitis, and biofilm formation on the implants in the surgical legs compared with sham-operated surgical legs without implant placement and with contralateral nonoperated normal legs. Neutralizing human monoclonal antibodies against α-toxin (AT) and clumping factor A (ClfA), especially in combination, inhibited biofilm formation in vitro and the hematogenous implant-related infection in vivo. Our findings suggest that AT and ClfA are pathogenic factors that could be therapeutically targeted against Saureus hematogenous implant-related infections.


Asunto(s)
Anticuerpos Antibacterianos/farmacología , Anticuerpos Neutralizantes/farmacología , Artritis Infecciosa , Biopelículas/efectos de los fármacos , Implantes Experimentales/microbiología , Osteomielitis , Infecciones Estafilocócicas , Staphylococcus aureus/fisiología , Animales , Artritis Infecciosa/tratamiento farmacológico , Artritis Infecciosa/etiología , Artritis Infecciosa/microbiología , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Osteomielitis/tratamiento farmacológico , Osteomielitis/etiología , Osteomielitis/microbiología , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/etiología , Infecciones Estafilocócicas/microbiología , Titanio
13.
J Allergy Clin Immunol ; 143(4): 1426-1443.e6, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30240702

RESUMEN

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


Asunto(s)
Dermatitis Atópica/metabolismo , Inflamación/metabolismo , Interleucina-1alfa/metabolismo , Proteínas de Filamentos Intermediarios/deficiencia , Queratinocitos/metabolismo , Animales , Dermatitis Atópica/inmunología , Dermatitis Atópica/microbiología , Disbiosis/inmunología , Disbiosis/metabolismo , Proteínas Filagrina , Inflamación/inmunología , Inflamación/microbiología , Interleucina-1alfa/inmunología , Queratinocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados
14.
Artículo en Inglés | MEDLINE | ID: mdl-31138566

RESUMEN

Surgical site infections (SSIs) are commonly caused by Staphylococcus aureus We report that a combination of three monoclonal antibodies (MEDI6389) that neutralize S. aureus alpha-toxin, clumping factor A, and four leukocidins (LukSF, LukED, HlgAB, and HlgCB) plus vancomycin had enhanced efficacy compared with control antibody plus vancomycin in two mouse models of S. aureus SSI. Therefore, monoclonal antibody-based neutralization of multiple S. aureus virulence factors may provide an adjunctive perioperative approach to combat S. aureus SSIs.


Asunto(s)
Antibacterianos/farmacología , Anticuerpos Monoclonales/farmacología , Infecciones Estafilocócicas/tratamiento farmacológico , Infección de la Herida Quirúrgica/tratamiento farmacológico , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Proteínas Bacterianas/inmunología , Anticuerpos ampliamente neutralizantes/farmacología , Coagulasa/inmunología , Leucocidinas/inmunología , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Ratones Endogámicos C57BL , Ratones Endogámicos , Infecciones Estafilocócicas/microbiología , Infección de la Herida Quirúrgica/microbiología , Vancomicina/farmacología
15.
J Immunol ; 199(5): 1772-1782, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28733486

RESUMEN

The immune response to Staphylococcus aureus infection in skin involves the recruitment of polymorphonuclear neutrophils (PMNs) from the bone marrow via the circulation and local granulopoiesis from hematopoietic stem and progenitor cells (HSPCs) that also traffic to infected skin wounds. We focus on regulation of PMN number and function and the role of pore-forming α-toxin (AT), a virulence factor that causes host cell lysis and elicits inflammasome-mediated IL-1ß secretion in wounds. Infection with wild-type S. aureus enriched in AT reduced PMN recruitment and resulted in sustained bacterial burden and delayed wound healing. In contrast, PMN recruitment to wounds infected with an isogenic AT-deficient S. aureus strain was unimpeded, exhibiting efficient bacterial clearance and hastened wound resolution. HSPCs recruited to infected wounds were unaffected by AT production and were activated to expand PMN numbers in proportion to S. aureus abundance in a manner regulated by TLR2 and IL-1R signaling. Immunodeficient MyD88-knockout mice infected with S. aureus experienced lethal sepsis that was reversed by PMN expansion mediated by injection of wild-type HSPCs directly into wounds. We conclude that AT-induced IL-1ß promotes local granulopoiesis and effective resolution of S. aureus-infected wounds, revealing a potential antibiotic-free strategy for tuning the innate immune response to treat methicillin-resistant S. aureus infection in immunodeficient patients.


Asunto(s)
Toxinas Bacterianas/inmunología , Granulocitos/inmunología , Células Madre Hematopoyéticas/fisiología , Proteínas Hemolisinas/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/fisiología , Factores de Virulencia/inmunología , Infección de Heridas/inmunología , Animales , Carga Bacteriana , Toxinas Bacterianas/genética , Diferenciación Celular , Proliferación Celular , Granulocitos/microbiología , Proteínas Hemolisinas/genética , Inmunomodulación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Factor 88 de Diferenciación Mieloide/genética , Receptores de Interleucina-1/metabolismo , Transducción de Señal , Receptor Toll-Like 2/metabolismo , Factores de Virulencia/genética
16.
Proc Natl Acad Sci U S A ; 113(45): E6919-E6928, 2016 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-27791154

RESUMEN

Bacterial biofilm formation is a major complication of implantable medical devices that results in therapeutically challenging chronic infections, especially in cases involving antibiotic-resistant bacteria. As an approach to prevent these infections, an electrospun composite coating comprised of poly(lactic-coglycolic acid) (PLGA) nanofibers embedded in a poly(ε-caprolactone) (PCL) film was developed to locally codeliver combinatorial antibiotics from the implant surface. The release of each antibiotic could be adjusted by loading each drug into the different polymers or by varying PLGA:PCL polymer ratios. In a mouse model of biofilm-associated orthopedic-implant infection, three different combinations of antibiotic-loaded coatings were highly effective in preventing infection of the bone/joint tissue and implant biofilm formation and were biocompatible with enhanced osseointegration. This nanofiber composite-coating technology could be used to tailor the delivery of combinatorial antimicrobial agents from various metallic implantable devices or prostheses to effectively decrease biofilm-associated infections in patients.

17.
Artículo en Inglés | MEDLINE | ID: mdl-29311091

RESUMEN

Staphylococcus aureus wound infections delay healing and result in invasive complications such as osteomyelitis, especially in the setting of diabetic foot ulcers. In preclinical animal models of S. aureus skin infection, antibody neutralization of alpha-toxin (AT), an S. aureus-secreted pore-forming cytolytic toxin, reduces disease severity by inhibiting skin necrosis and restoring effective host immune responses. However, whether therapeutic neutralization of alpha-toxin is effective against S. aureus-infected wounds is unclear. Herein, the efficacy of prophylactic treatment with a human neutralizing anti-AT monoclonal antibody (MAb) was evaluated in an S. aureus skin wound infection model in nondiabetic and diabetic mice. In both nondiabetic and diabetic mice, anti-AT MAb treatment decreased wound size and bacterial burden and enhanced reepithelialization and wound resolution compared to control MAb treatment. Anti-AT MAb had distinctive effects on the host immune response, including decreased neutrophil and increased monocyte and macrophage infiltrates in nondiabetic mice and decreased neutrophil extracellular traps (NETs) in diabetic mice. Similar therapeutic efficacy was achieved with an active vaccine targeting AT. Taken together, neutralization of AT had a therapeutic effect against S. aureus-infected wounds in both nondiabetic and diabetic mice that was associated with differential effects on the host immune response.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Toxinas Bacterianas/antagonistas & inhibidores , Diabetes Mellitus Experimental/inmunología , Proteínas Hemolisinas/antagonistas & inhibidores , Infecciones Cutáneas Estafilocócicas/tratamiento farmacológico , Cicatrización de Heridas/efectos de los fármacos , Heridas no Penetrantes/tratamiento farmacológico , Animales , Carga Bacteriana/efectos de los fármacos , Toxinas Bacterianas/inmunología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/microbiología , Trampas Extracelulares/efectos de los fármacos , Trampas Extracelulares/microbiología , Proteínas Hemolisinas/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Monocitos/microbiología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/microbiología , Piel/efectos de los fármacos , Piel/inmunología , Piel/microbiología , Infecciones Cutáneas Estafilocócicas/complicaciones , Infecciones Cutáneas Estafilocócicas/inmunología , Infecciones Cutáneas Estafilocócicas/microbiología , Vacunas Estafilocócicas/farmacología , Cicatrización de Heridas/inmunología , Heridas no Penetrantes/complicaciones , Heridas no Penetrantes/inmunología , Heridas no Penetrantes/microbiología
18.
Infect Immun ; 85(2)2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27849182

RESUMEN

Staphylococcus aureus is the leading cause of skin and skin structure infections (SSSI). The high frequency of recurring SSSI due to S. aureus, including methicillin-resistant S. aureus (MRSA) strains, despite high titers of specific antibodies and circulating T cells, implies that traditional adaptive immunity imparts incomplete protection. We hypothesized that innate immune memory contributes to the protective host defense against recurring MRSA infection. To test this hypothesis, SSSI was induced in wild-type and rag1-/- mice in the BALB/c and C57BL/6 backgrounds. Prior infection (priming) of wild-type and rag1-/- mice of either background afforded protection against repeat infection, as evidenced by reduced abscess severities and decreased CFU densities compared to those in naive controls. Interestingly, protection was greater on the previously infected flank than on the naive flank for wild-type and rag1-/- mice. For wild-type mice, protective efficacy corresponded to increased infiltration of neutrophils (polymorphonuclear leukocytes [PMN]), macrophages (MΦ), Langerin+ dendritic cells (LDC), and natural killer (NK) cells. Protection was associated with the induction of interleukin-17A (IL-17A), IL-22, and gamma interferon (IFN-γ) as well as the antimicrobial peptides CRAMP and mßD-3. Priming also protected rag1-/- mice against recurring SSSI, with increased MΦ and LDC infiltration and induction of IL-22, CRAMP, and mßD-3. These findings suggest that innate immune memory, mediated by specific cellular and molecular programs, likely contributes to the localized host defense in recurrent MRSA SSSI. These insights support the development of targeted immunotherapeutic strategies to address the challenge of MRSA infection.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Memoria Inmunológica , Staphylococcus aureus Resistente a Meticilina/inmunología , Infecciones Cutáneas Estafilocócicas/inmunología , Infecciones Cutáneas Estafilocócicas/microbiología , Animales , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Expresión Génica , Masculino , Ratones , Ratones Noqueados , Recurrencia , Bazo/citología , Bazo/inmunología , Infecciones Cutáneas Estafilocócicas/patología
19.
Am J Pathol ; 186(9): 2337-52, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27470712

RESUMEN

Infections with Staphylococcus aureus are a continuing and growing problem in community and hospital settings. Preclinical animal modeling of S. aureus relies on experimental infection, which carries some limitations. We describe here a novel, spontaneous model of oral staphylococcal infection in double knockout mice, deficient in the receptors for IL-17 (IL-17RA) and interferon (IFN)-γ (IFNγRI), beginning at 6 to 8 weeks of age. IFNγRI(-/-)IL17RA(-/-) (GRAKO) mice developed progressive oral abscesses. Cytometric methods revealed extensive neutrophilic infiltration of oral tissues in GRAKO mice; further investigation evidenced that IL-17 predominated neutrophil defects in these mice. To investigate the contribution of IFN-γ signaling to this native host defense to S. aureus, we observed perturbations of monocyte recruitment and macrophage differentiation in the oral tissues of GRAKO mice, and CXCL9/chemokine ligand receptor (CXCR)3-driven recruitment of T-cell oral tissues and draining lymph nodes. To address the former finding, we depleted macrophages and monocytes in vivo from IL17RA(-/-) mice using liposomes loaded with clodronate. This treatment elicited oral abscesses, recapitulating the phenotype of GRAKO mice. From these findings, we propose novel collaborative functions of IL-17 and IFN-γ, acting through neutrophils and macrophages, respectively, in native mucocutaneous host defenses to S. aureus.


Asunto(s)
Interferón gamma/inmunología , Interleucina-17/inmunología , Mucosa Bucal/inmunología , Mucosa Bucal/microbiología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Transducción de Señal/inmunología
20.
J Allergy Clin Immunol ; 138(4): 1098-1107.e3, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27543072

RESUMEN

BACKGROUND: Staphylococcus aureus is an opportunistic pathogen that colonizes the skin of patients with atopic dermatitis (AD) and aggravates their disease. Neutrophils and the cytokines IL-17A and IL-17F, which drive the expression of the neutrophil-attracting chemokines, are important for the clearance of S aureus infection. The cytokine IL-22 is often coproduced by IL-17-secreting cells. The levels of IL-22 are elevated in AD skin lesions. OBJECTIVE: We sought to determine the role of IL-22 in the clearance of S aureus infection of mouse skin subjected to tape stripping, a surrogate for scratching, a cardinal feature of AD. METHODS: S aureus was applied to the tape-stripped skin of wild-type and Il22-/- mice. Bacterial burden was evaluated by enumerating colony-forming units. Quantitative PCR and ELISA were performed to quantify Il22 mRNA and IL-22 protein in mouse and human skin. Flow cytometry was used to enumerate neutrophils in the skin. RESULTS: Scratching the skin of healthy adults and tape stripping of mouse skin induced local expression of Il22 mRNA and IL-22 protein. Induction of Il22 expression by tape stripping was dependent on IL-23 and γδ T cells. Clearance of S aureus from tape-stripped skin was significantly impaired in Il22-/- mice. Neutrophil infiltration and upregulation of expression of genes encoding the antimicrobial peptides antigen-6/urokinase-type plasminogen activator receptor related protein-1 and ß-DEFENSIN 14 and the chemokine (C-X-C motif) ligand following tape stripping were significantly impaired in Il22-/- mice. CONCLUSIONS: These findings show that IL-22 is important for limiting the growth of S aureus on mechanically injured skin and caution that IL-23 and IL-22 blockade in patients with AD may enhance susceptibility to staphylococcal skin infection.


Asunto(s)
Interleucinas/farmacología , Infecciones Cutáneas Estafilocócicas/prevención & control , Staphylococcus aureus/efectos de los fármacos , Linfocitos T/inmunología , Animales , Dermatitis Atópica/inmunología , Citometría de Flujo , Humanos , Interleucinas/aislamiento & purificación , Ratones , Piel/lesiones , Piel/microbiología , Linfocitos T/química , Interleucina-22
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA