Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Immunity ; 50(2): 446-461.e9, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30709742

RESUMEN

Production of interleukin-17 (IL-17) and IL-22 by T helper 17 (Th17) cells and group 3 innate lymphoid cells (ILC3s) in response to the gut microbiota ensures maintenance of intestinal barrier function. Here, we examined the mechanisms whereby the immune system detects microbiota in the steady state. A Syk-kinase-coupled signaling pathway in dendritic cells (DCs) was critical for commensal-dependent production of IL-17 and IL-22 by CD4+ T cells. The Syk-coupled C-type lectin receptor Mincle detected mucosal-resident commensals in the Peyer's patches (PPs), triggered IL-6 and IL-23p19 expression, and thereby regulated function of intestinal Th17- and IL-17-secreting ILCs. Mice deficient in Mincle or with selective depletion of Syk in CD11c+ cells had impaired production of intestinal RegIIIγ and IgA and increased systemic translocation of gut microbiota. Consequently, Mincle deficiency led to liver inflammation and deregulated lipid metabolism. Thus, sensing of commensals by Mincle and Syk signaling in CD11c+ cells reinforces intestinal immune barrier and promotes host-microbiota mutualism, preventing systemic inflammation.


Asunto(s)
Células Dendríticas/inmunología , Microbioma Gastrointestinal/inmunología , Interleucina-17/inmunología , Interleucinas/inmunología , Lectinas Tipo C/inmunología , Proteínas de la Membrana/inmunología , Quinasa Syk/inmunología , Animales , Células Dendríticas/metabolismo , Microbioma Gastrointestinal/fisiología , Humanos , Interleucina-17/metabolismo , Interleucinas/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/metabolismo , Ganglios Linfáticos Agregados/microbiología , Transducción de Señal/inmunología , Quinasa Syk/genética , Quinasa Syk/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Interleucina-22
2.
Annu Rev Microbiol ; 76: 757-782, 2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-36075093

RESUMEN

Identified in the late nineteenth century as a single species residing on human skin, Malassezia is now recognized as a diverse genus comprising 18 species inhabiting not only skin but human gut, hospital environments, and even deep-sea sponges. All cultivated Malassezia species are lipid dependent, having lost genes for lipid synthesis and carbohydrate metabolism. The surging interest in Malassezia results from development of tools to improve sampling, culture, identification, and genetic engineering, which has led to findings implicating it in numerous skin diseases, Crohn disease, and pancreatic cancer. However, it has become clear that Malassezia plays a multifaceted role in human health, with mutualistic activity in atopic dermatitis and a preventive effect against other skin infections due to its potential to compete with skin pathogens such as Candida auris. Improved understanding of complex microbe-microbe and host-microbe interactions will be required to define Malassezia's role in human and animal health and disease so as to design targeted interventions.


Asunto(s)
Dermatitis Atópica , Malassezia , Animales , Humanos , Lípidos , Malassezia/genética , Piel , Simbiosis
3.
PLoS Pathog ; 20(1): e1011668, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38215167

RESUMEN

Stable microbial colonization of the skin depends on tight control by the host immune system. The lipid-dependent yeast Malassezia typically colonizes skin as a harmless commensal and is subject to host type 17 immunosurveillance, but this fungus has also been associated with diverse skin pathologies in both humans and animals. Using a murine model of Malassezia exposure, we show that Vγ4+ dermal γδ T cells expand rapidly and are the major source of IL-17A mediating fungal control in colonized skin. A pool of memory-like Malassezia-responsive Vγ4+ T cells persisted in the skin, were enriched in draining lymph nodes even after fungal clearance, and were protective upon fungal re-exposure up to several weeks later. Induction of γδT17 immunity depended on IL-23 and IL-1 family cytokine signalling, whereas Toll-like and C-type lectin receptors were dispensable. Furthermore, Vγ4+ T cells from Malassezia-exposed hosts were able to respond directly and selectively to Malassezia-derived ligands, independently of antigen-presenting host cells. The fungal moieties detected were shared across diverse species of the Malassezia genus, but not conserved in other Basidiomycota or Ascomycota. These data provide novel mechanistic insight into the induction and maintenance of type 17 immunosurveillance of skin commensal colonization that has significant implications for cutaneous health.


Asunto(s)
Malassezia , Humanos , Ratones , Animales , Saccharomyces cerevisiae , Interleucina-17 , Linfocitos T , Alérgenos
4.
Proc Natl Acad Sci U S A ; 119(49): e2212533119, 2022 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-36442106

RESUMEN

Malassezia form the dominant eukaryotic microbial community on the human skin. The Malassezia genus possesses a repertoire of secretory hydrolytic enzymes involved in protein and lipid metabolism which alter the external cutaneous environment. The exact role of most Malassezia secreted enzymes, including those in interaction with the epithelial surface, is not well characterized. In this study, we compared the expression level of secreted proteases, lipases, phospholipases, and sphingomyelinases of Malassezia globosa in healthy subjects and seborrheic dermatitis or atopic dermatitis patients. We observed upregulated gene expression of the previously characterized secretory aspartyl protease MGSAP1 in both diseased groups, in lesional and non-lesional skin sites, as compared to healthy subjects. To explore the functional roles of MGSAP1 in skin disease, we generated a knockout mutant of the homologous protease MFSAP1 in the genetically tractable Malassezia furfur. We observed the loss of MFSAP1 resulted in dramatic changes in the cell adhesion and dispersal in both culture and a human 3D reconstituted epidermis model. In a murine model of Malassezia colonization, we further demonstrated Mfsap1 contributes to inflammation as observed by reduced edema and inflammatory cell infiltration with the knockout mutant versus wildtype. Taken together, we show that this dominant secretory Malassezia aspartyl protease has an important role in enabling a planktonic cellular state that can potentially aid in colonization and additionally as a virulence factor in barrier-compromised skin, further highlighting the importance of considering the contextual relevance when evaluating the functions of secreted microbial enzymes.


Asunto(s)
Proteasas de Ácido Aspártico , Dermatitis Atópica , Malassezia , Humanos , Animales , Ratones , Péptido Hidrolasas/genética , Malassezia/genética , Inflamación , Ácido Aspártico Endopeptidasas
5.
PLoS Pathog ; 18(4): e1010012, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35404986

RESUMEN

As part of the human microbiota, the fungus Candida albicans colonizes the oral cavity and other mucosal surfaces of the human body. Commensalism is tightly controlled by complex interactions of the fungus and the host to preclude fungal elimination but also fungal overgrowth and invasion, which can result in disease. As such, defects in antifungal T cell immunity render individuals susceptible to oral thrush due to interrupted immunosurveillance of the oral mucosa. The factors that promote commensalism and ensure persistence of C. albicans in a fully immunocompetent host remain less clear. Using an experimental model of C. albicans oral colonization in mice we explored fungal determinants of commensalism in the oral cavity. Transcript profiling of the oral isolate 101 in the murine tongue tissue revealed a characteristic metabolic profile tailored to the nutrient poor conditions in the stratum corneum of the epithelium where the fungus resides. Metabolic adaptation of isolate 101 was also reflected in enhanced nutrient acquisition when grown on oral mucosa substrates. Persistent colonization of the oral mucosa by C. albicans also correlated inversely with the capacity of the fungus to induce epithelial cell damage and to elicit an inflammatory response. Here we show that these immune evasive properties of isolate 101 are explained by a strong attenuation of a number of virulence genes, including those linked to filamentation. De-repression of the hyphal program by deletion or conditional repression of NRG1 abolished the commensal behaviour of isolate 101, thereby establishing a central role of this factor in the commensal lifestyle of C. albicans in the oral niche of the host.


Asunto(s)
Candida albicans , Candidiasis Bucal , Animales , Candidiasis Bucal/microbiología , Proteínas Fúngicas , Ratones , Mucosa Bucal/microbiología , Simbiosis , Virulencia
6.
Allergy ; 79(6): 1531-1547, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38385963

RESUMEN

BACKGROUND: The skin barrier is vital for protection against environmental threats including insults caused by skin-resident microbes. Dysregulation of this barrier is a hallmark of atopic dermatitis (AD) and ichthyosis, with variable consequences for host immune control of colonizing commensals and opportunistic pathogens. While Malassezia is the most abundant commensal fungus of the skin, little is known about the host control of this fungus in inflammatory skin diseases. METHODS: In this experimental study, MC903-treated mice were colonized with Malassezia spp. to assess the host-fungal interactions in atopic dermatitis. Additional murine models of AD and ichthyosis, including tape stripping, K5-Nrf2 overexpression and flaky tail mice, were employed to confirm and expand the findings. Skin fungal counts were enumerated. High parameter flow cytometry was used to characterize the antifungal response in the AD-like skin. Structural and functional alterations in the skin barrier were determined by histology and transcriptomics of bulk skin. Finally, differential expression of metabolic genes in Malassezia in atopic and control skin was quantified. RESULTS: Malassezia grows excessively in AD-like skin. Fungal overgrowth could, however, not be explained by the altered immune status of the atopic skin. Instead, we found that by upregulating key metabolic genes in the altered cutaneous niche, Malassezia acquired enhanced fitness to efficiently colonise the impaired skin barrier. CONCLUSIONS: This study provides evidence that structural and metabolic changes in the dysfunctional epidermal barrier environment provide increased accessibility and an altered lipid profile, to which the lipid-dependent yeast adapts for enhanced nutrient assimilation. Our findings reveal fundamental insights into the implication of the mycobiota in the pathogenesis of common skin barrier disorders.


Asunto(s)
Dermatitis Atópica , Modelos Animales de Enfermedad , Malassezia , Piel , Animales , Malassezia/inmunología , Ratones , Dermatitis Atópica/microbiología , Dermatitis Atópica/inmunología , Piel/microbiología , Piel/inmunología , Epidermis/microbiología , Epidermis/inmunología , Epidermis/metabolismo , Susceptibilidad a Enfermedades , Hipersensibilidad/inmunología , Hipersensibilidad/microbiología , Femenino
7.
Immunity ; 40(1): 117-27, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24412614

RESUMEN

Interleukin 17 (IL-17)-mediated immunity plays a key role in protection from fungal infections in mice and man. Here, we confirmed that mice deficient in the IL-17 receptor or lacking the ability to secrete IL-17 are highly susceptible to systemic candidiasis, but we found that temporary blockade of the IL-17 pathway during infection in wild-type mice did not impact fungal control. Rather, mice lacking IL-17 receptor signaling had a cell-intrinsic impairment in the development of functional NK cells, which accounted for the susceptibility of these mice to systemic fungal infection. NK cells promoted antifungal immunity by secreting GM-CSF, necessary for the fungicidal activity of neutrophils. These data reveal that NK cells are crucial for antifungal defense and indicate a role for IL-17 family cytokines in NK cell development. The IL-17-NK cell axis may impact immunity against not only fungi but also bacteria, viruses, and tumors.


Asunto(s)
Candidiasis/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Células Asesinas Naturales/inmunología , Neutrófilos/inmunología , Receptores de Interleucina-17/metabolismo , Animales , Candidiasis/genética , Diferenciación Celular , Células Cultivadas , Citotoxicidad Inmunológica , Susceptibilidad a Enfermedades , Interleucina-17/genética , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Interleucina-17/genética , Transducción de Señal/genética
8.
Parasite Immunol ; 45(2): e12948, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36047038

RESUMEN

Fungi represent an integral part of the skin microbiota. Their complex interaction network with the host shapes protective immunity during homeostasis. If host defences are breached, skin-resident fungi including Malassezia and Candida, and environmental fungi such as dermatophytes can cause cutaneous infections. In addition, fungi are associated with diverse non-infectious skin disorders. Despite their multiple roles in health and disease, fungi remain elusive and understudied, and the mechanisms underlying the emergence of pathological conditions linked to fungi are largely unclear. The identification of IL-17 as an important antifungal effector mechanism represents a milestone for understanding homeostatic antifungal immunity. At the same time, host-adverse, disease-promoting roles of IL-17 have been delineated, as in psoriasis. Fungal dysbiosis represents another feature of many pathological skin conditions with an unknown causal link of intra- and interkingdom interactions to disease pathogenesis. The emergence of new fungal pathogens such as Candida auris highlights the need for more research into fungal immunology to understand how antifungal responses shape health and diseases. Recent technological advances for genetically manipulating fungi to target immunomodulatory fungal determinants, multi-omics approaches for studying immune cells in the human skin, and novel experimental models open up a promising future for skin fungal immunity.


Asunto(s)
Malassezia , Microbiota , Humanos , Interleucina-17 , Antifúngicos , Piel , Hongos/fisiología
9.
Proc Natl Acad Sci U S A ; 117(27): 15884-15894, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32576698

RESUMEN

The skin of humans and animals is colonized by commensal and pathogenic fungi and bacteria that share this ecological niche and have established microbial interactions. Malassezia are the most abundant fungal skin inhabitant of warm-blooded animals and have been implicated in skin diseases and systemic disorders, including Crohn's disease and pancreatic cancer. Flavohemoglobin is a key enzyme involved in microbial nitrosative stress resistance and nitric oxide degradation. Comparative genomics and phylogenetic analyses within the Malassezia genus revealed that flavohemoglobin-encoding genes were acquired through independent horizontal gene transfer events from different donor bacteria that are part of the mammalian microbiome. Through targeted gene deletion and functional complementation in Malassezia sympodialis, we demonstrated that bacterially derived flavohemoglobins are cytoplasmic proteins required for nitric oxide detoxification and nitrosative stress resistance under aerobic conditions. RNA-sequencing analysis revealed that endogenous accumulation of nitric oxide resulted in up-regulation of genes involved in stress response and down-regulation of the MalaS7 allergen-encoding genes. Solution of the high-resolution X-ray crystal structure of Malassezia flavohemoglobin revealed features conserved with both bacterial and fungal flavohemoglobins. In vivo pathogenesis is independent of Malassezia flavohemoglobin. Lastly, we identified an additional 30 genus- and species-specific horizontal gene transfer candidates that might have contributed to the evolution of this genus as the most common inhabitants of animal skin.


Asunto(s)
Bacterias/genética , Hemoproteínas/genética , Interacciones Microbiota-Huesped/fisiología , Malassezia/genética , Malassezia/metabolismo , Óxido Nítrico/metabolismo , Piel/microbiología , Animales , Bacterias/metabolismo , Cristalografía por Rayos X , Ergosterol/biosíntesis , Evolución Molecular , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Eliminación de Gen , Regulación Fúngica de la Expresión Génica , Transferencia de Gen Horizontal , Hemoproteínas/química , Hemoproteínas/metabolismo , Humanos , Malassezia/clasificación , Modelos Moleculares , Estrés Oxidativo/genética , Estrés Oxidativo/fisiología , Filogenia , Piel/metabolismo , Simbiosis
10.
J Am Chem Soc ; 143(27): 10389-10402, 2021 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-34212720

RESUMEN

Mutanobactin D is a non-ribosomal, cyclic peptide isolated from Streptococcus mutans and shows activity reducing yeast-to-hyphae transition as well as biofilm formation of the pathogenic yeast Candida albicans. We report the first total synthesis of this natural product, which relies on enantioselective, zinc-mediated 1,3-dipolar cycloaddition and a sequence of cascading reactions, providing the key lipidated γ-amino acid found in mutanobactin D. The synthesis enables configurational assignment, determination of the dominant solution-state structure, and studies to assess the stability of the lipopeptide substructure found in the natural product. The information stored in the fingerprint region of the IR spectra in combination with quantum chemical calculations proved key to distinguishing between epimers of the α-substituted ß-keto amide. Synthetic mutanobactin D drives discovery and analysis of its effect on growth of other members of the human oral consortium. Our results showcase how total synthesis is central for elucidating the complex network of interspecies communications of human colonizers.


Asunto(s)
Antifúngicos/farmacología , Péptidos Cíclicos , Antifúngicos/química , Candida albicans/efectos de los fármacos , Hifa/efectos de los fármacos , Modelos Moleculares , Péptidos Cíclicos/síntesis química , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología
11.
PLoS Pathog ; 15(12): e1008115, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31887131

RESUMEN

The opportunistic fungal pathogen Candida albicans can cause invasive infections in susceptible hosts and the innate immune system, in particular myeloid cell-mediated immunity, is critical for rapid immune protection and host survival during systemic candidiasis. Using a mouse model of the human disease, we identified a novel role of IL-23 in antifungal defense. IL-23-deficient mice are highly susceptible to systemic infection with C. albicans. We found that this results from a drastic reduction in all subsets of myeloid cells in the infected kidney, which in turn leads to rapid fungal overgrowth and renal tissue injury. The loss in myeloid cells is not due to a defect in emergency myelopoiesis or the recruitment of newly generated cells to the site of infection but, rather, is a consequence of impaired survival of myeloid cells at the site of infection. In fact, the absence of a functional IL-23 pathway causes massive myeloid cell apoptosis upon C. albicans infection. Importantly, IL-23 protects myeloid cells from apoptosis independently of the IL-23-IL-17 immune axis and independently of lymphocytes and innate lymphoid cells. Instead, our results suggest that IL-23 acts in a partially autocrine but not cell-intrinsic manner within the myeloid compartment to promote host protection from systemic candidiasis. Collectively, our data highlight an unprecedented and non-canonical role of IL-23 in securing survival of myeloid cells, which is key for maintaining sufficient numbers of cells at the site of infection to ensure efficient host protection.


Asunto(s)
Candida albicans/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Interleucina-23/farmacología , Células Progenitoras Mieloides/efectos de los fármacos , Animales , Candida albicans/inmunología , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Interleucina-17/metabolismo , Interleucina-23/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Ratones Endogámicos C57BL , Células Mieloides/metabolismo
12.
Parasitology ; 148(1): 53-62, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33087186

RESUMEN

Susceptibility to Echinococcus multilocularis infection considerably varies among intermediate (mostly rodents) and dead-end host species (e.g. humans and pig), in particular regarding intestinal oncosphere invasion and subsequent hepatic metacestode development. Wistar rats are highly resistant to infection and subsequent diseases upon oral inoculation with E. multilocularis eggs, however, after immunosuppressive treatment with dexamethasone, rats become susceptible. To address the role of the cellular innate immunity, Wistar rats were individually or combined depleted of natural killer (NK) cells, macrophages (MΦ) and granulocytes (polymorphonuclear cells, PMN) prior to E. multilocularis egg inoculation. Although NK cell and MΦ depletion did not alter the resistance status of rats, the majority of PMN-depleted animals developed liver metacestodes within 10 weeks, indicating that PMN are key players in preventing oncosphere migration and/or development in Wistar rats. In vitro studies indicated that resistance is not caused by neutrophil reactive oxygen species or NETosis. Also, light microscopical examinations of the small intestine showed that oral inoculation of E. multilocularis eggs does not elicit a mucosal neutrophil response, suggesting that the interaction of oncospheres and neutrophils may occur after the former have entered the peripheral blood. We suggest to consider granulocytes as mediators of resistance in more resistant species, such as humans.


Asunto(s)
Agranulocitosis/complicaciones , Equinococosis Hepática/inmunología , Echinococcus multilocularis , Inmunidad Innata , Animales , Modelos Animales de Enfermedad , Resistencia a la Enfermedad , Susceptibilidad a Enfermedades/inducido químicamente , Equinococosis/inmunología , Echinococcus multilocularis/crecimiento & desarrollo , Echinococcus multilocularis/inmunología , Granulocitos/inmunología , Inmunidad Mucosa , Inmunosupresores/administración & dosificación , Intestinos/inmunología , Intestinos/parasitología , Células Asesinas Naturales/inmunología , Hígado/parasitología , Macrófagos/inmunología , Neutrófilos/inmunología , Ratas , Ratas Wistar/parasitología
13.
PLoS Pathog ; 14(5): e1007069, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29782555

RESUMEN

The opportunistic fungal pathogen Candida albicans frequently causes diseases such as oropharyngeal candidiasis (OPC) in immunocompromised individuals. Although it is well appreciated that the cytokine IL-17 is crucial for protective immunity against OPC, the cellular source and the regulation of this cytokine during infection are still a matter of debate. Here, we directly visualized IL-17 production in the tongue of experimentally infected mice, thereby demonstrating that this key cytokine is expressed by three complementary subsets of CD90+ leukocytes: RAG-dependent αß and γδ T cells, as well as RAG-independent ILCs. To determine the regulation of IL-17 production at the onset of OPC, we investigated in detail the myeloid compartment of the tongue and found a heterogeneous and dynamic mononuclear phagocyte (MNP) network in the infected tongue that consists of Zbtb46-Langerin- macrophages, Zbtb46+Langerin+ dendritic cells (DCs) and Ly6C+ inflammatory monocytes. Of those, the Langerin+ DC population stands out by its unique capacity to co-produce the cytokines IL-1ß, IL-6 and IL-23, all of which promote IL-17 induction in response to C. albicans in the oral mucosa. The critical role of Langerin+ DCs for the innate IL-17 response was confirmed by depletion of this cellular subset in vivo, which compromised IL-17 induction during OPC. In conclusion, our work revealed key regulatory factors and their cellular sources of innate IL-17-dependent antifungal immunity in the oral mucosa.


Asunto(s)
Antígenos de Superficie/inmunología , Candida albicans/inmunología , Candidiasis Bucal/inmunología , Células Dendríticas/inmunología , Interleucina-17/biosíntesis , Lectinas Tipo C/inmunología , Lectinas de Unión a Manosa/inmunología , Mucosa Bucal/inmunología , Animales , Candidiasis Bucal/microbiología , Citocinas/inmunología , Femenino , Citometría de Flujo , Interleucina-1beta/biosíntesis , Interleucina-23/biosíntesis , Interleucina-23/inmunología , Interleucina-6/biosíntesis , Leucocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Sistema Mononuclear Fagocítico/inmunología , Mucosa Bucal/citología , Mucosa Bucal/microbiología , Neutrófilos/inmunología , Organismos Libres de Patógenos Específicos , Bazo/citología , Bazo/inmunología , Antígenos Thy-1/inmunología , Lengua/citología , Lengua/inmunología , Lengua/microbiología
15.
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
16.
Int J Mol Sci ; 22(1)2020 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-33375121

RESUMEN

Regulatory T cells (Tregs) prevent excessive immune responses and limit immune pathology upon infections. To fulfill this role in different immune environments elicited by different types of pathogens, Tregs undergo functional specialization into distinct subsets. During acute type 1 immune responses, type 1 Tregs are induced and recruited to the site of ongoing Th1 responses to efficiently control Th1 responses. However, whether a similar specialization process also takes place following chronic infections is still unknown. In this study, we investigated Treg specialization in persistent viral infections using lymphocytic choriomeningitis virus (LCMV) and murine cytomegalovirus (MCMV) infection as models for chronic and latent infections, respectively. We identify CD85k as a Th1-specific co-inhibitory receptor with sustained expression in persistent viral infections and show that recombinant CD85k inhibits LCMV-specific effector T cells. Furthermore, expression of the CD85k ligand ALCAM is induced on LCMV-specific and exhausted T cells during chronic LCMV infection. Finally, we demonstrate that type 1 Tregs arising during chronic LCMV infection suppress Th1 effector cells in an ALCAM-dependent manner. These results extend the current knowledge of Treg specialization from acute to persistent viral infections and reveal an important functional role of CD85k in Treg-mediated suppression of type 1 immunity.


Asunto(s)
Virus de la Coriomeningitis Linfocítica/inmunología , Glicoproteínas de Membrana/inmunología , Muromegalovirus/inmunología , Receptores Inmunológicos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígenos CD/inmunología , Antígenos CD/metabolismo , Moléculas de Adhesión Celular Neuronal/inmunología , Moléculas de Adhesión Celular Neuronal/metabolismo , Línea Celular , Células Cultivadas , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/virología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/metabolismo , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/fisiología , Glicoproteínas de Membrana/metabolismo , Ratones Endogámicos C57BL , Muromegalovirus/fisiología , Receptores Inmunológicos/metabolismo , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/virología , Células TH1/inmunología , Células TH1/metabolismo
17.
Cell Microbiol ; 20(8): e12841, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29575428

RESUMEN

The fungus Candida albicans thrives on a variety of human mucosae, yet the fungal determinants that contribute to fitness on these surfaces remain underexplored. Here, by screening a collection of C. albicans deletion strains in a mouse model of oral infection (oropharyngeal candidiasis), we identify several novel regulatory genes that modulate the fitness of the fungus in this locale. We investigate in detail the interplay between the host mucosa and one of the identified mutants and establish that the C. albicans transcription regulator CUP9 is a key determinant of mucosal colonisation. Deletion of cup9 resulted in the formation of more foci of colonisation and heightened persistence in infected tongues. Furthermore, the cup9 mutant produced longer and denser filaments in the oral mucosa without eliciting an enhanced local immune response. Consistent with its role in oral colonisation, we show that CUP9's top target of regulation is a major effector of Candida's adherence to buccal cells. Finally, we establish that CUP9 also governs the interplay of the fungus with vaginal epithelial cells and has a role in vaginal infections, another common mucosal disease associated with Candida. Thus, our findings reveal a mechanism whereby C. albicans can regulate proliferation on mucosal surfaces.


Asunto(s)
Candida albicans/genética , Candida albicans/patogenicidad , Candidiasis Bucal/microbiología , Candidiasis Vulvovaginal/microbiología , Genes Reguladores , Membrana Mucosa/microbiología , Factores de Transcripción/metabolismo , Animales , Candida albicans/crecimiento & desarrollo , Adhesión Celular , Modelos Animales de Enfermedad , Células Epiteliales/microbiología , Femenino , Eliminación de Gen , Pruebas Genéticas , Interacciones Huésped-Patógeno , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Factores de Transcripción/genética , Vagina/microbiología , Virulencia
18.
Mol Microbiol ; 106(1): 157-182, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28752552

RESUMEN

Skn7 is a conserved fungal heat shock factor-type transcriptional regulator. It participates in maintaining cell wall integrity and regulates the osmotic/oxidative stress response (OSR) in S. cerevisiae, where it is part of a two-component signal transduction system. Here, we comprehensively address the function of Skn7 in the human fungal pathogen Candida albicans. We provide evidence reinforcing functional divergence, with loss of the cell wall/osmotic stress-protective roles and acquisition of the ability to regulate morphogenesis on solid medium. Mapping of the Skn7 transcriptional circuitry, through combination of genome-wide expression and location technologies, pointed to a dual regulatory role encompassing OSR and filamentous growth. Genetic interaction analyses revealed close functional interactions between Skn7 and master regulators of morphogenesis, including Efg1, Cph1 and Ume6. Intracellular biochemical assays revealed that Skn7 is crucial for limiting the accumulation of reactive oxygen species (ROS) in filament-inducing conditions on solid medium. Interestingly, functional domain mapping using site-directed mutagenesis allowed decoupling of Skn7 function in morphogenesis from protection against intracellular ROS. Our work identifies Skn7 as an integral part of the transcriptional circuitry controlling C. albicans filamentous growth and illuminates how C. albicans relies on an evolutionarily-conserved regulator to protect itself from intracellular ROS during morphological development.


Asunto(s)
Candida albicans/crecimiento & desarrollo , Candida albicans/genética , Candida albicans/metabolismo , Pared Celular/metabolismo , Secuencia Conservada/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Fúngicas/metabolismo , Regulación Fúngica de la Expresión Génica/genética , Genes Fúngicos/genética , Hifa/crecimiento & desarrollo , Morfogénesis , Especies Reactivas de Oxígeno/metabolismo , Elementos de Respuesta/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Análisis de Secuencia de ADN , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Transcripción Genética/genética
19.
PLoS Pathog ; 12(9): e1005882, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27632536

RESUMEN

Mucosal infections with Candida albicans belong to the most frequent forms of fungal diseases. Host protection is conferred by cellular immunity; however, the induction of antifungal immunity is not well understood. Using a mouse model of oropharyngeal candidiasis (OPC) we show that interleukin-1 receptor (IL-1R) signaling is critical for fungal control at the onset of infection through its impact on neutrophils at two levels. We demonstrate that both the recruitment of circulating neutrophils to the site of infection and the mobilization of newly generated neutrophils from the bone marrow depended on IL-1R. Consistently, IL-1R-deficient mice displayed impaired chemokine production at the site of infection and defective secretion of granulocyte colony-stimulating factor (G-CSF) in the circulation in response to C. albicans. Strikingly, endothelial cells were identified as the primary cellular source of G-CSF during OPC, which responded to IL-1α that was released from keratinocytes in the infected tissue. The IL-1-dependent crosstalk between two different cellular subsets of the nonhematopoietic compartment was confirmed in vitro using a novel murine tongue-derived keratinocyte cell line and an established endothelial cell line. These data establish a new link between IL-1 and granulopoiesis in the context of fungal infection. Together, we identified two complementary mechanisms coordinating the neutrophil response in the oral mucosa, which is critical for preventing fungal growth and dissemination, and thus protects the host from disease.


Asunto(s)
Candida albicans/inmunología , Candidiasis/inmunología , Interleucina-1alfa/inmunología , Mucosa Bucal/inmunología , Neutrófilos/inmunología , Animales , Candidiasis/genética , Células Endoteliales/inmunología , Células Endoteliales/microbiología , Factor Estimulante de Colonias de Granulocitos/genética , Factor Estimulante de Colonias de Granulocitos/inmunología , Interleucina-1alfa/genética , Queratinocitos/inmunología , Queratinocitos/microbiología , Ratones , Ratones Noqueados , Mucosa Bucal/microbiología , Mielopoyesis/genética , Mielopoyesis/inmunología , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/inmunología , Lengua/inmunología , Lengua/microbiología
20.
PLoS Pathog ; 12(6): e1005723, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27341123

RESUMEN

Salmonella Typhimurium (S.Tm) is a common cause of self-limiting diarrhea. The mucosal inflammation is thought to arise from a standoff between the pathogen's virulence factors and the host's mucosal innate immune defenses, particularly the mucosal NAIP/NLRC4 inflammasome. However, it had remained unclear how this switches the gut from homeostasis to inflammation. This was studied using the streptomycin mouse model. S.Tm infections in knockout mice, cytokine inhibition and -injection experiments revealed that caspase-1 (not -11) dependent IL-18 is pivotal for inducing acute inflammation. IL-18 boosted NK cell chemoattractants and enhanced the NK cells' migratory capacity, thus promoting mucosal accumulation of mature, activated NK cells. NK cell depletion and Prf-/- ablation (but not granulocyte-depletion or T-cell deficiency) delayed tissue inflammation. Our data suggest an NK cell perforin response as one limiting factor in mounting gut mucosal inflammation. Thus, IL-18-elicited NK cell perforin responses seem to be critical for coordinating mucosal inflammation during early infection, when S.Tm strongly relies on virulence factors detectable by the inflammasome. This may have broad relevance for mucosal defense against microbial pathogens.


Asunto(s)
Interleucina-18/biosíntesis , Mucosa Intestinal/inmunología , Células Asesinas Naturales/inmunología , Proteínas Citotóxicas Formadoras de Poros/inmunología , Infecciones por Salmonella/inmunología , Animales , Caspasa 1 , Quimiotaxis de Leucocito/inmunología , Modelos Animales de Enfermedad , Citometría de Flujo , Inmunidad Innata/inmunología , Inflamasomas/inmunología , Inflamación/inmunología , Interleucina-18/inmunología , Cinética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Reacción en Cadena de la Polimerasa , Salmonella typhimurium/inmunología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda