Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
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.
Nat Immunol ; 19(9): 1001-1012, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30104633

RESUMEN

Immunoglobulin G3 (IgG3) has an uncertain role in the response to infection with and vaccination against human immunodeficiency virus (HIV). Here we describe a regulatory role for IgG3 in dampening the immune system-activating effects of chronic HIV viremia on B cells. Secreted IgG3 was bound to IgM-expressing B cells in vivo in HIV-infected chronically viremic individuals but not in early-viremic or aviremic individuals. Tissue-like memory (TLM) B cells, a population expanded by persistent HIV viremia, bound large amounts of IgG3. IgG3 induced clustering of B cell antigen receptors (BCRs) on the IgM+ B cells, which was mediated by direct interactions between soluble IgG3 and membrane IgM of the BCR (IgM-BCR). The inhibitory IgG receptor CD32b (FcγRIIb), complement component C1q and inflammatory biomarker CRP contributed to the binding of secreted IgG3 onto IgM-expressing B cells of HIV-infected individuals. Notably, IgG3-bound TLM B cells were refractory to IgM-BCR stimulation, thus demonstrating that IgG3 can regulate B cells during chronic activation of the immune system.


Asunto(s)
Linfocitos B/inmunología , Infecciones por VIH/inmunología , VIH-1/fisiología , Inmunoglobulina G/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Adulto , Proteína C-Reactiva/metabolismo , Células Cultivadas , Complemento C1q/metabolismo , Femenino , Humanos , Inmunoglobulina M/metabolismo , Memoria Inmunológica , Inmunomodulación , Masculino , Persona de Mediana Edad , Unión Proteica , Agregación de Receptores , Receptores de IgG/metabolismo , Adulto Joven
3.
Proc Natl Acad Sci U S A ; 121(6): e2309243121, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38289950

RESUMEN

Staphylococcus aureus skin colonization and eosinophil infiltration are associated with many inflammatory skin disorders, including atopic dermatitis, bullous pemphigoid, Netherton's syndrome, and prurigo nodularis. However, whether there is a relationship between S. aureus and eosinophils and how this interaction influences skin inflammation is largely undefined. We show in a preclinical mouse model that S. aureus epicutaneous exposure induced eosinophil-recruiting chemokines and eosinophil infiltration into the skin. Remarkably, we found that eosinophils had a comparable contribution to the skin inflammation as T cells, in a manner dependent on eosinophil-derived IL-17A and IL-17F production. Importantly, IL-36R signaling induced CCL7-mediated eosinophil recruitment to the inflamed skin. Last, S. aureus proteases induced IL-36α expression in keratinocytes, which promoted infiltration of IL-17-producing eosinophils. Collectively, we uncovered a mechanism for S. aureus proteases to trigger eosinophil-mediated skin inflammation, which has implications in the pathogenesis of inflammatory skin diseases.


Asunto(s)
Dermatitis Atópica , Eosinofilia , Infecciones Estafilocócicas , Animales , Ratones , Eosinófilos/metabolismo , Staphylococcus aureus/metabolismo , Péptido Hidrolasas/metabolismo , Piel/metabolismo , Dermatitis Atópica/metabolismo , Infecciones Estafilocócicas/metabolismo , Celulitis (Flemón)/metabolismo , Celulitis (Flemón)/patología , Inflamación/metabolismo
4.
EMBO Rep ; 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38783164

RESUMEN

Neutrophil extracellular traps (NETs) are a key antimicrobial feature of cellular innate immunity mediated by polymorphonuclear neutrophils (PMNs). NETs counteract microbes but are also linked to inflammation in atherosclerosis, arthritis, or psoriasis by unknown mechanisms. Here, we report that NET-associated RNA (naRNA) stimulates further NET formation in naive PMNs via a unique TLR8-NLRP3 inflammasome-dependent pathway. Keratinocytes respond to naRNA with expression of psoriasis-related genes (e.g., IL17, IL36) via atypical NOD2-RIPK signaling. In vivo, naRNA drives temporary skin inflammation, which is drastically ameliorated by genetic ablation of RNA sensing. Unexpectedly, the naRNA-LL37 'composite damage-associated molecular pattern (DAMP)' is pre-stored in resting neutrophil granules, defining sterile NETs as inflammatory webs that amplify neutrophil activation. However, the activity of the naRNA-LL37 DAMP is transient and hence supposedly self-limiting under physiological conditions. Collectively, upon dysregulated NET release like in psoriasis, naRNA sensing may represent both a potential cause of disease and a new intervention target.

5.
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
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.
Int J Mol Sci ; 18(8)2017 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-28813021

RESUMEN

The multipotency and anti-inflammatory effects of mesenchymal stem cells (MSCs) make them attractive for cell therapy in regenerative medicine. A large number of MSCs is required for efficient therapy owing to the low homing efficiency of MSCs to target sites. Furthermore, owing to limitations in obtaining sufficient amounts of MSCs, in vitro expansion of MSCs that preserves their differentiation and proliferative potential is essential. The animal factor included in culture media also limits clinical application. In this study, adipose-derived MSCs showed a significantly higher proliferation rate in STK2, a chemically-defined medium, than in DMEM/FBS. The expression of MSC surface markers was increased in the culture using STK2 compared to that using DMEM/FBS. Tri-lineage differentiation analyses showed that MSCs cultured in STK2 were superior to those cultured in DMEM/FBS. In addition, MSCs cultured in STK2 showed a reduced senescence rate, small and homogenous cell size, and were more genetically stable compared to those cultured in DMEM/FBS. Furthermore, secretome analysis showed that the expression of factors related to proliferation/migration, anti-inflammation, and differentiation were increased in STK2 culture medium compared to DMEM/FBS. Taken together, these results suggest that culture using STK2 medium offers many advantages through which it is possible to obtain safer, superior, and larger numbers of MSCs.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Técnicas de Cultivo de Célula/métodos , Medios de Cultivo/química , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Humanos
8.
Front Immunol ; 14: 1171934, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37483624

RESUMEN

Staphylococcus aureus is a leading cause of bacteremia, further complicated by the emergence of antibiotic-resistant strains such as methicillin-resistant S. aureus (MRSA). A better understanding of host defense mechanisms is needed for the development of host-directed therapies as an alternative approach to antibiotics. The levels of IL-1, IL-17, and TNF-α cytokines in circulation have been associated with predictive outcomes in patients with S. aureus bacteremia. However, their causative role in survival and the cell types involved in these responses during bacteremia is not entirely clear. Using a mouse model of S. aureus bacteremia, we demonstrated that IL-17A/F and TNF-α had no significant impact on survival, whereas IL-1R signaling was critical for survival during S. aureus bacteremia. Furthermore, we identified that T cells, but not neutrophils, monocytes/macrophages, or endothelial cells were the crucial cell type for IL-1R-mediated survival against S. aureus bacteremia. Finally, we determined that the expression of IL-1R on γδ T cell, but not CD4+ or CD8+ T cells was responsible for survival against the S. aureus bacteremia. Taken together, we uncovered a role for IL-1R, but not IL-17A/F and TNF-α in protection against S. aureus bacteremia. Importantly, γδ T cell-intrinsic expression of IL-1R was crucial for survival, but not on other immune cells or endothelial cells. These findings reveal potential cellular and immunological targets for host-directed therapies for improved outcomes against S. aureus bacteremia.


Asunto(s)
Bacteriemia , Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Humanos , Staphylococcus aureus , Staphylococcus aureus Resistente a Meticilina/fisiología , Factor de Necrosis Tumoral alfa , Linfocitos T CD8-positivos , Células Endoteliales , Bacteriemia/prevención & control
9.
Sci Adv ; 9(24): eadf8748, 2023 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-37327341

RESUMEN

Staphylococcus aureus is the leading cause of skin and soft tissue infections and is a major health burden due to the emergence of antibiotic-resistant strains. To address the unmet need of alternative treatments to antibiotics, a better understanding of the protective immune mechanisms against S. aureus skin infection is warranted. Here, we report that tumor necrosis factor (TNF) promoted protection against S. aureus in the skin, which was mediated by bone marrow-derived immune cells. Furthermore, neutrophil-intrinsic TNF receptor (TNFR) signaling directed immunity against S. aureus skin infections. Mechanistically, TNFR1 promoted neutrophil recruitment to the skin, whereas TNFR2 prevented systemic bacterial dissemination and directed neutrophil antimicrobial functions. Treatment with a TNFR2 agonist showed therapeutic efficacy against S. aureus and Pseudomonas aeruginosa skin infections, which involved increased neutrophil extracellular trap formation. Our findings revealed nonredundant roles for TNFR1 and TNFR2 in neutrophils for immunity against S. aureus and can be therapeutically targeted for protection against bacterial skin infections.


Asunto(s)
Neutrófilos , Infecciones Estafilocócicas , Humanos , Staphylococcus aureus , Receptores Tipo I de Factores de Necrosis Tumoral , Receptores Tipo II del Factor de Necrosis Tumoral , Infecciones Estafilocócicas/tratamiento farmacológico
10.
J Invest Dermatol ; 142(4): 1126-1135.e4, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34626614

RESUMEN

Signal transducer and activator of transcription 3 (STAT3) is important for psoriasis pathogenesis because STAT3 signaling downstream of IL-6, IL-21, IL-22, and IL-23 contributes to T helper type 17 cell development and because transgenic mice with keratinocyte (KC) STAT3 expression (K14-Stat3C mice) develop psoriasis-like dermatitis. In this study, the relative contribution of STAT3 signaling in KCs versus in T cells was evaluated in the imiquimod model of psoriasis-like dermatitis. Mice with STAT3-inducible deletion in KCs (K5-Stat3-/- mice) had decreased psoriasis-like dermatitis and epidermal STAT3 phosphorylation compared with wild-type mice, whereas mice with constitutive deletion of STAT3 in all T cells were similar to wild-type mice. Interestingly, mice with KC-inducible deletion of IL-6Rα had similar findings to those of K5-Stat3-/- mice, identifying IL-6/IL-6R as a predominant upstream signal for KC STAT3-induced psoriasis-like dermatitis. Moreover, psoriasis-like dermatitis inversely associated with type 1 immune gene products, especially CXCL10, whereas CXCL10 limited psoriasis-like dermatitis, suggesting that KC STAT3 signaling promoted psoriasis-like dermatitis by restricting downstream CXCL10 expression. Finally, treatment of mice with the pan-Jak inhibitor, tofacitinib, reduced psoriasis-like dermatitis and epidermal STAT3 phosphorylation. Taken together, STAT3 signaling in KCs rather than in T cells was a more important determinant for psoriasis-like dermatitis in a mechanism that involved upstream KC IL-6R signaling and downstream inhibition of type 1 immunity‒associated CXCL10 responses.


Asunto(s)
Dermatitis , Psoriasis , Animales , Quimiocina CXCL10 , Dermatitis/patología , Modelos Animales de Enfermedad , Interleucina-6/metabolismo , Queratinocitos/metabolismo , Ratones , Ratones Transgénicos , Receptores de Interleucina-6 , Factor de Transcripción STAT3/metabolismo , Linfocitos T/metabolismo
11.
Microbiol Spectr ; 10(5): e0245121, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36106881

RESUMEN

Staphylococcus aureus is an important cause of various infections in humans, including bacteremia, skin and soft tissue infections, and infections associated with implanted medical devices. The emergence of hospital- and community-acquired methicillin-resistant Staphylococcus aureus (MRSA) underscores the urgent and unmet need to develop novel, safe, and effective antibiotics against these multidrug-resistant clinical isolates. Oxazolidinone antibiotics such as linezolid have excellent oral bioavailability and provide coverage against MRSA infections. However, their widespread and long-term use is often limited by adverse effects, especially myelosuppression. TBI-223 is a novel oxazolidinone with potentially reduced myelosuppression, compared to linezolid, but its efficacy against MRSA infections is unknown. Therefore, the preclinical efficacy of TBI-223 (80 and 160 mg/kg twice daily) was compared with that of linezolid (40 and 80 mg/kg twice daily) and sham treatment in mouse models of MRSA bacteremia, skin wound infection, and orthopedic-implant-associated infection. The dosage was selected based on mouse pharmacokinetic analysis of both linezolid and TBI-223, as well as measurement of the MICs. In all three models, TBI-223 and linezolid had comparable dose-dependent efficacies in reducing bacterial burden and disease severity, compared with sham-treated control mice. Taken together, these findings indicate that TBI-223 represents a novel oxazolidinone antibiotic that may provide an additional option against MRSA infections. Future studies in larger animal models and clinical trials are warranted to translate these findings to humans. IMPORTANCE Staphylococcus aureus is the predominant cause of bloodstream, skin, and bone infections in humans. Resistance to commonly used antibiotics is a growing concern, making it more difficult to treat staphylococcal infections. Use of the oxazolidinone antibiotic linezolid against resistant strains is hindered by high rates of adverse reactions during prolonged therapy. Here, a new oxazolidinone named TBI-223 was tested against S. aureus in three mouse models of infection, i.e., bloodstream infection, skin infection, and bone infection. We found that TBI-223 was as effective as linezolid in these three models. Previous data suggest that TBI-223 has a better safety profile than linezolid. Taken together, these findings indicate that this new agent may provide an additional option against MRSA infections. Future studies in larger animal models and clinical trials are warranted to translate these findings to humans.


Asunto(s)
Bacteriemia , Staphylococcus aureus Resistente a Meticilina , Oxazolidinonas , Infecciones Estafilocócicas , Animales , Ratones , Acetamidas/farmacología , Acetamidas/uso terapéutico , Antibacterianos/efectos adversos , Bacteriemia/tratamiento farmacológico , Linezolid/efectos adversos , Pruebas de Sensibilidad Microbiana , Oxazolidinonas/efectos adversos , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus
12.
Sci Transl Med ; 13(601)2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-34233954

RESUMEN

Staphylococcus aureus causes most skin infections in humans, and the emergence of methicillin-resistant S. aureus (MRSA) strains is a serious public health threat. There is an urgent clinical need for nonantibiotic immunotherapies to treat MRSA infections and prevent the spread of antibiotic resistance. Here, we investigated the pan-caspase inhibitor quinoline-valine-aspartic acid-difluorophenoxymethyl ketone (Q-VD-OPH) for efficacy against MRSA skin infection in mice. A single systemic dose of Q-VD-OPH decreased skin lesion sizes and reduced bacterial burden compared with vehicle-treated or untreated mice. Although Q-VD-OPH inhibited inflammasome-dependent apoptosis-associated speck-like protein containing caspase activation and recruitment domain (ASC) speck formation and caspase-1-mediated interleukin-1ß (IL-1ß) production, Q-VD-OPH maintained efficacy in mice deficient in IL-1ß, ASC, caspase-1, caspase-11, or gasdermin D. Thus, Q-VD-OPH efficacy was independent of inflammasome-mediated pyroptosis. Rather, Q-VD-OPH reduced apoptosis of monocytes and neutrophils. Moreover, Q-VD-OPH enhanced necroptosis of macrophages with concomitant increases in serum TNF and TNF-producing neutrophils, monocytes/macrophages, and neutrophils in the infected skin. Consistent with this, Q-VD-OPH lacked efficacy in mice deficient in TNF (with associated reduced neutrophil influx and necroptosis), in mice deficient in TNF/IL-1R and anti-TNF antibody-treated WT mice. In vitro studies revealed that combined caspase-3, caspase-8, and caspase-9 inhibition reduced apoptosis, and combined caspase-1, caspase-8, and caspase-11 inhibition increased TNF, suggesting a mechanism for Q-VD-OPH efficacy in vivo. Last, Q-VD-OPH also had a therapeutic effect against Streptococcus pyogenes and Pseudomonas aeruginosa skin infections in mice. Collectively, pan-caspase inhibition represents a potential host-directed immunotherapy against MRSA and other bacterial skin infections.


Asunto(s)
Caspasas , Staphylococcus aureus Resistente a Meticilina , Animales , Caspasa 1 , Inhibidores de Caspasas/farmacología , Inmunoterapia , Inflamasomas , Interleucina-1beta , Ratones , Inhibidores del Factor de Necrosis Tumoral
13.
J Clin Invest ; 131(5)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33645549

RESUMEN

IgE induced by type 2 immune responses in atopic dermatitis is implicated in the progression of atopic dermatitis to other allergic diseases, including food allergies, allergic rhinitis, and asthma. However, the keratinocyte-derived signals that promote IgE and ensuing allergic diseases remain unclear. Herein, in a mouse model of atopic dermatitis-like skin inflammation induced by epicutaneous Staphylococcus aureus exposure, keratinocyte release of IL­36α along with IL-4 triggered B cell IgE class-switching, plasma cell differentiation, and increased serum IgE levels-all of which were abrogated in IL-36R-deficient mice or anti-IL­36R-blocking antibody-treated mice. Moreover, skin allergen sensitization during S. aureus epicutaneous exposure-induced IL-36 responses was required for the development of allergen-specific lung inflammation. In translating these findings, elevated IL­36 cytokines in human atopic dermatitis skin and in IL­36 receptor antagonist-deficiency patients coincided with increased serum IgE levels. Collectively, keratinocyte-initiated IL­36 responses represent a key mechanism and potential therapeutic target against allergic diseases.


Asunto(s)
Dermatitis Atópica/inmunología , Inmunoglobulina E/inmunología , Interleucina-1/inmunología , Queratinocitos/inmunología , Células Plasmáticas/inmunología , Staphylococcus aureus/inmunología , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Dermatitis Atópica/genética , Dermatitis Atópica/microbiología , Humanos , Cambio de Clase de Inmunoglobulina , Inmunoglobulina E/genética , Interleucina-1/genética , Interleucina-4/genética , Interleucina-4/inmunología , Queratinocitos/microbiología , Ratones , Ratones Noqueados , Células Plasmáticas/patología
14.
J Invest Dermatol ; 140(8): 1488-1497.e1, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32407714

RESUMEN

Bacterial skin infections are a major societal health burden and are increasingly difficult to treat owing to the emergence of antibiotic-resistant strains such as community-acquired methicillin-resistant Staphylococcus aureus. Understanding the immunologic mechanisms that provide durable protection against skin infections has the potential to guide the development of immunotherapies and vaccines to engage the host immune response to combat these antibiotic-resistant strains. To this end, mouse skin infection models allow researchers to examine host immunity by investigating the timing, inoculum, route of infection and the causative bacterial species in different wild-type mouse backgrounds as well as in knockout, transgenic, and other types of genetically engineered mouse strains. To recapitulate the various types of human skin infections, many different mouse models have been developed. For example, four models frequently used in dermatological research are based on the route of infection, including (i) subcutaneous infection models, (ii) intradermal infection models, (iii) wound infection models, and (iv) epicutaneous infection models. In this article, we will describe these skin infection models in detail along with their advantages and limitations. In addition, we will discuss how humanized mouse models such as the human skin xenograft on immunocompromised mice might be used in bacterial skin infection research.


Asunto(s)
Modelos Animales de Enfermedad , Huésped Inmunocomprometido , Staphylococcus aureus Resistente a Meticilina/inmunología , Piel/microbiología , Infecciones Cutáneas Estafilocócicas/inmunología , Animales , Infecciones Comunitarias Adquiridas/inmunología , Infecciones Comunitarias Adquiridas/microbiología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proyectos de Investigación , Piel/inmunología , Trasplante de Piel , Infecciones Cutáneas Estafilocócicas/microbiología , Quimera por Trasplante
15.
J Clin Invest ; 130(7): 3805-3819, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32298242

RESUMEN

Microbial ingestion by a macrophage results in the formation of an acidic phagolysosome but the host cell has no information on the pH susceptibility of the ingested organism. This poses a problem for the macrophage and raises the fundamental question of how the phagocytic cell optimizes the acidification process to prevail. We analyzed the dynamical distribution of phagolysosomal pH in murine and human macrophages that had ingested live or dead Cryptococcus neoformans cells, or inert beads. Phagolysosomal acidification produced a range of pH values that approximated normal distributions, but these differed from normality depending on ingested particle type. Analysis of the increments of pH reduction revealed no forbidden ordinal patterns, implying that the phagosomal acidification process was a stochastic dynamical system. Using simulation modeling, we determined that by stochastically acidifying a phagolysosome to a pH within the observed distribution, macrophages sacrificed a small amount of overall fitness to gain the benefit of reduced variation in fitness. Hence, chance in the final phagosomal pH introduces unpredictability to the outcome of the macrophage-microbe, which implies a bet-hedging strategy that benefits the macrophage. While bet hedging is common in biological systems at the organism level, our results show its use at the organelle and cellular level.


Asunto(s)
Criptococosis/inmunología , Cryptococcus neoformans/inmunología , Macrófagos/inmunología , Fagosomas/inmunología , Animales , Línea Celular , Femenino , Humanos , Concentración de Iones de Hidrógeno , Ratones
16.
PLoS One ; 9(9): e107000, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25188500

RESUMEN

The steroid hormone receptors regulate important physiological functions such as reproduction, metabolism, immunity, and electrolyte balance. Mutations within steroid receptors result in endocrine disorders and can often drive cancer formation and progression. Despite the conserved three-dimensional structure shared among members of the steroid receptor family and their overlapping DNA binding preference, activation of individual steroid receptors drive unique effects on gene expression. Here, we present the first structure of the human mineralocorticoid receptor DNA binding domain, in complex with a canonical DNA response element. The overall structure is similar to the glucocorticoid receptor DNA binding domain, but small changes in the mode of DNA binding and lever arm conformation may begin to explain the differential effects on gene regulation by the mineralocorticoid and glucocorticoid receptors. In addition, we explore the structural effects of mineralocorticoid receptor DNA binding domain mutations found in type I pseudohypoaldosteronism and multiple types of cancer.


Asunto(s)
ADN/química , Receptores de Glucocorticoides/química , Receptores de Mineralocorticoides/química , Secuencia de Aminoácidos , Cristalografía por Rayos X , ADN/genética , ADN/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Humanos , Datos de Secuencia Molecular , Mutación , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Seudohipoaldosteronismo/genética , Seudohipoaldosteronismo/metabolismo , Seudohipoaldosteronismo/patología , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/genética , Receptores de Mineralocorticoides/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología Estructural de Proteína
17.
Nat Struct Mol Biol ; 20(1): 53-8, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23222642

RESUMEN

A newly discovered negative glucocorticoid response element (nGRE) mediates DNA-dependent transrepression by the glucocorticoid receptor (GR) across the genome and has a major role in immunosuppressive therapy. The nGRE differs dramatically from activating response elements, and the mechanism driving GR binding and transrepression is unknown. To unravel the mechanism of nGRE-mediated transrepression by the GR, we characterized the interaction between GR and an nGRE in the thymic stromal lymphopoietin (TSLP) promoter. We show using structural and mechanistic approaches that nGRE binding is a new mode of sequence recognition by human GR and that nGREs prevent receptor dimerization through a unique GR-binding orientation and strong negative cooperativity, ensuring the presence of monomeric GR at repressive elements.


Asunto(s)
Citocinas/genética , ADN/metabolismo , Glucocorticoides/metabolismo , Receptores de Glucocorticoides/metabolismo , Elementos de Respuesta , Sitios de Unión , Línea Celular , Proteínas de Unión al ADN/metabolismo , Glucocorticoides/genética , Células HeLa , Humanos , Mutación , Regiones Promotoras Genéticas , Unión Proteica , Multimerización de Proteína , Receptores de Glucocorticoides/química , Receptores de Glucocorticoides/genética , Secuencias Reguladoras de Ácidos Nucleicos , Transcripción Genética , Linfopoyetina del Estroma Tímico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA