Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-38260364

RESUMEN

Aspergillus fumigatus causes life-threatening mold pneumonia in immune compromised patients, particularly in those with quantitative or qualitative defects in neutrophils. While innate immune cell crosstalk licenses neutrophil antifungal activity in the lung, the role of epithelial cells in this process is unknown. Here, we find that that surfactant protein C (SPC)-expressing lung epithelial cells integrate infection-induced IL-1 and type III interferon signaling to produce granulocyte-macrophage colony-stimulating factor (GM-CSF) preferentially at local sites of fungal infection and neutrophil influx. Using in vivo models that distinguish the role of GM-CSF during acute infection from its homeostatic function in alveolar macrophage survival and surfactant catabolism, we demonstrate that epithelial-derived GM-CSF increases the accumulation and fungicidal activity of GM-CSF-responsive neutrophils, with the latter being essential for host survival. Our findings establish SPC + epithelial cells as a central player in regulating the quality and strength of neutrophil-dependent immunity against inhaled mold pathogens. HIGHLIGHTS: GM-CSF is essential for host defense against A. fumigatus in the lung IL-1 and IFN-λ promote GM-CSF production by lung epithelial cells in parallelEpithelial cell-derived GM-CSF increases neutrophil accumulation and fungal killing capacityEpithelial cells preferentially upregulate GM-CSF in local sites of inflammation.

2.
PNAS Nexus ; 2(11): pgad350, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37954158

RESUMEN

Interferon ɛ (IFNɛ) is a unique type I IFN that has been implicated in host defense against sexually transmitted infections. Zika virus (ZIKV), an emerging pathogen, can infect the female reproductive tract (FRT) and cause devastating diseases, particularly in pregnant women. How IFNɛ contributes to protection against ZIKV infection in vivo is unknown. In this study, we show that IFNɛ plays a critical role in host protection against vaginal ZIKV infection in mice. We found that IFNɛ was expressed not only by epithelial cells in the FRT but also by immune and stromal cells at baseline or after exposure to viruses or specific Toll-like receptor (TLR) agonists. IFNɛ-deficient mice exhibited abnormalities in the epithelial border and underlying tissue in the cervicovaginal tract, and these defects were associated with increased susceptibility to vaginal but not subcutaneous ZIKV infection. IFNɛ deficiency resulted in an increase in magnitude, duration, and depth of ZIKV infection in the FRT. Critically, intravaginal administration of recombinant IFNɛ protected Ifnɛ-/- mice and highly susceptible Ifnar1-/- mice against vaginal ZIKV infection, indicating that IFNɛ was sufficient to provide protection even in the absence of signals from other type I IFNs and in an IFNAR1-independent manner. Our findings reveal a potentially critical role for IFNɛ in mediating protection against the transmission of ZIKV in the context of sexual contact.

3.
Curr Opin Immunol ; 84: 102371, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37523967

RESUMEN

The detrimental impact of fungal infections to human health has steadily increased over the past decades. In October of 2022, the World Health Organization published the first ever fungal-pathogen priority list highlighting increased awareness of this problem, and the need for more research in this area. There were four distinct fungal pathogens identified as critical priority groups with Aspergillus fumigatus (Af) being the only mold. Af is a common environmental fungus responsible for over 90% of invasive aspergillosis cases worldwide. Pulmonary protection against Af is critically dependent on innate effector cells with essential roles played by neutrophils and monocytes. In this review, we will summarize our current understanding of how monocytes help orchestrate antifungal defense against Af.


Asunto(s)
Aspergilosis , Monocitos , Humanos , Aspergillus fumigatus , Neutrófilos , Pulmón
4.
bioRxiv ; 2023 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-37066223

RESUMEN

Interferon ε (IFNε) is a unique type I IFN that has been implicated in host defense against sexually transmitted infections (STIs). Zika virus (ZIKV), an emerging pathogen, can infect the female reproductive tract (FRT) and cause devastating diseases, particularly in pregnant women. How IFNε contributes to protection against ZIKV infection in vivo is unknown. Here, we show that IFNε plays a critical role in host protection against vaginal ZIKV infection in mice. We found that IFNε was expressed not only by epithelial cells in the FRT, but also by certain immune and other cells at baseline or after exposure to viruses or specific TLR agonists. IFNε-deficient mice exhibited abnormalities in the epithelial border and underlying tissue in the cervicovaginal tract, and these defects were associated with increased susceptibility to vaginal, but not subcutaneous ZIKV infection. IFNε-deficiency resulted in an increase in magnitude, duration, and depth of ZIKV infection in the FRT. Critically, intravaginal administration of recombinant IFNε protected Ifnε-/- mice and highly susceptible Ifnar1-/- mice against vaginal ZIKV infection, indicating that IFNε was sufficient to provide protection even in the absence of signals from other type I IFNs and in an IFNAR1-independent manner. Our findings reveal a potentially critical role for IFNε in mediating protection against transmission of ZIKV in the context of sexual contact.

5.
Semin Immunol ; 66: 101728, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36841146

RESUMEN

The respiratory tree maintains sterilizing immunity against human fungal pathogens. Humans inhale ubiquitous filamentous molds and geographically restricted dimorphic fungal pathogens that form small airborne conidia. In addition, pathogenic yeasts, exemplified by encapsulated Cryptococcus species, and Pneumocystis pose significant fungal threats to the lung. Classically, fungal pneumonia occurs in immune compromised individuals, specifically in patients with HIV/AIDS, in patients with hematologic malignancies, in organ transplant recipients, and in patients treated with corticosteroids and targeted biologics that impair fungal immune surveillance in the lung. The emergence of fungal co-infections during severe influenza and COVID-19 underscores the impairment of fungus-specific host defense pathways in the lung by respiratory viruses and by medical therapies to treat viral infections. Beyond life-threatening invasive syndromes, fungal antigen exposure can exacerbate allergenic disease in the lung. In this review, we discuss emerging principles of lung-specific antifungal immunity, integrate the contributions and cooperation of lung epithelial, innate immune, and adaptive immune cells to mucosal barrier immunity, and highlight the pathogenesis of fungal-associated allergenic disease. Improved understanding of fungus-specific immunity in the respiratory tree has paved the way to develop improved diagnostic, pre-emptive, therapeutic, and vaccine approaches for fungal diseases of the lung.


Asunto(s)
COVID-19 , Micosis , Humanos , Pulmón , Hongos , Inmunidad Innata
6.
Microbiol Spectr ; : e0495522, 2023 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-36719231

RESUMEN

In previous studies, we determined that the F-box protein Fbp1, a subunit of the SCF(Fbp1) E3 ligase in Cryptococcus neoformans, is essential for fungal pathogenesis. Heat-killed fbp1Δ cells (HK-fbp1) can confer vaccine-induced immunity against lethal challenge with clinically important invasive fungal pathogens, e.g., C. neoformans, C. gattii, and Aspergillus fumigatus. In this study, we found that either CD4+ T cells or CD8+ T cells were sufficient to confer protection against lethal challenge by C. neoformans in HK-fbp1-induced immunity. Given the potent effect of HK-fbp1 as a preventative vaccine, we further tested the potential efficacy of administering HK-fbp1 cells as a therapeutic agent for treating animals after infection. Remarkably, administration of HK-fbp1 provided robust host protection against preexisting C. neoformans infection. The mice infected with wild-type H99 cells and then treated with HK-fbp1 showed significant reduction of fungal burden in the infected lung and no dissemination of fungal cells to the brain and spleen. We find that early treatment is critical for the effective use of HK-fbp1 as a therapeutic agent. Immune analysis revealed that early treatment with HK-fbp1 cells elicited Th1-biased protective immune responses that help block fungal dissemination and promote better host protection. Our data thus suggest that HK-fbp1 is both an effective prophylactic vaccine candidate against C. neoformans infection in both immunocompetent and immunocompromised populations and a potential novel therapeutic strategy to treat early-stage cryptococcosis. IMPORTANCE Invasive fungal infections, e.g., cryptococcosis, are often life threatening and difficult to treat with very limited therapeutic options. There is no vaccine available in clinical use to prevent or treat fungal infections. Our previous studies demonstrated that heat-killed fbp1Δ cells (HK-fbp1) in Cryptococcus neoformans can be harnessed to confer protection against a challenge by the virulent parental strain, even in immunocompromised animals, such as ones lacking CD4+ T cells. In this study, we further determined that T cells are required for vaccine-induced protection against homologous challenge and that either CD4+ or CD8+ cells are sufficient. This finding is particularly important for the potential utility of this vaccine candidate in the context of HIV/AIDS-induced immune deficiency, the main risk factor for cryptococcosis in humans. Furthermore, in addition to the utility of HK-fbp1 as a prophylactic vaccine, we found that HK-fbp1 administration can inhibit disease dissemination when animals are treated at an early stage during Cryptococcus infection. Our findings could significantly expand the utility of HK-fbp1 not only as a prophylactic vaccine but also as a novel therapy against cryptococcosis. In all, our studies showed that the HK-fbp1 strain can be used both preventively and therapeutically to elicit robust host protection against cryptococcosis.

7.
Nat Commun ; 13(1): 6397, 2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-36302775

RESUMEN

Fungal pathogens often undergo morphological switches, including cell size changes, to adapt to the host environment and cause disease. The pathogenic yeast Cryptococcus neoformans forms so-called 'titan cells' during infection. Titan cells are large, polyploid, display alterations in cell wall and capsule, and are more resistant to phagocytosis and various types of stress. Titan cell formation is regulated by the cAMP/PKA signal pathway, which is stimulated by the protein Gpa1. Here, we show that Gpa1 is activated through phosphorylation by a CDK-related kinase (Crk1), which is targeted for degradation by an E3 ubiquitin ligase (Fbp1). Strains overexpressing CRK1 or an allele lacking a PEST domain exhibit increased production of titan cells similarly to the fbp1∆ mutant. Conversely, CRK1 deletion results in reduced titan cell production, indicating that Crk1 stimulates titan cell formation. Crk1 phosphorylates Gpa1, which then localizes to the plasma membrane and activates the cAMP/PKA signal pathway to induce cell enlargement. Furthermore, titan cell-overproducing strains trigger increased Th1 and Th17 cytokine production in CD4+ T cells and show attenuated virulence in a mouse model of systemic cryptococcosis. Overall, our study provides insights into the regulation of titan cell formation and fungal virulence.


Asunto(s)
Criptococosis , Cryptococcus neoformans , Ratones , Animales , Virulencia , Ubiquitina/metabolismo , Proteolisis , Fosforilación , Criptococosis/microbiología , Saccharomyces cerevisiae/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo
8.
J Immunol ; 209(10): 1827-1831, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36216513

RESUMEN

Neutrophils are critical for the direct eradication of Aspergillus fumigatus conidia, but whether they mediate antifungal defense beyond their role as effectors is unclear. In this study, we demonstrate that neutrophil depletion impairs the activation of protective antifungal CCR2+ inflammatory monocytes. In the absence of neutrophils, monocytes displayed limited differentiation into monocyte-derived dendritic cells, reduced formation of reactive oxygen species, and diminished conidiacidal activity. Upstream regulator analysis of the transcriptional response in monocytes predicted a loss of STAT1-dependent signals as the potential basis for the dysfunction seen in neutrophil-depleted mice. We find that conditional removal of STAT1 on CCR2+ cells results in diminished antifungal monocyte responses, whereas exogenous administration of IFN-γ to neutrophil-depleted mice restores monocyte-derived dendritic cell maturation and reactive oxygen species production. Altogether, our findings support a critical role for neutrophils in antifungal immunity not only as effectors but also as important contributors to antifungal monocyte activation, in part by regulating STAT1-dependent functions.


Asunto(s)
Monocitos , Neutrófilos , Ratones , Animales , Antifúngicos , Especies Reactivas de Oxígeno , Aspergillus fumigatus
9.
Proc Natl Acad Sci U S A ; 119(37): e2201645119, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-36070344

RESUMEN

Neuroimmune interactions are crucial for regulating immunity and inflammation. Recent studies have revealed that the central nervous system (CNS) senses peripheral inflammation and responds by releasing molecules that limit immune cell activation, thereby promoting tolerance and tissue integrity. However, the extent to which this is a bidirectional process, and whether peripheral immune cells also promote tolerance mechanisms in the CNS remains poorly defined. Here we report that helminth-induced type 2 inflammation promotes monocyte responses in the brain that are required to inhibit excessive microglial activation and host death. Mechanistically, infection-induced monocytes express YM1 that is sufficient to inhibit tumor necrosis factor production from activated microglia. Importantly, neuroprotective monocytes persist in the brain, and infected mice are protected from subsequent lipopolysaccharide-induced neuroinflammation months after infection-induced inflammation has resolved. These studies demonstrate that infiltrating monocytes promote CNS homeostasis in response to inflammation in the periphery and demonstrate that a peripheral infection can alter the immunologic landscape of the host brain.


Asunto(s)
Encéfalo , Encefalitis , Homeostasis , Monocitos , Neuroinmunomodulación , Trichinella spiralis , Triquinelosis , Animales , Encéfalo/inmunología , Encéfalo/parasitología , Encefalitis/inmunología , Encefalitis/parasitología , Homeostasis/inmunología , Lectinas/metabolismo , Ratones , Microglía/inmunología , Monocitos/inmunología , Trichinella spiralis/inmunología , Triquinelosis/inmunología , Triquinelosis/patología , beta-N-Acetilhexosaminidasas/metabolismo
10.
J Immunol ; 209(4): 675-683, 2022 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-35879099

RESUMEN

Plasmacytoid dendritic cells (pDCs) have been implicated as having a role in antifungal immunity, but mechanisms of their interaction with fungi and the resulting cellular responses are not well understood. In this study, we identify the direct and indirect biological response of human pDCs to the fungal pathogen Aspergillus fumigatus and characterize the expression and regulation of antifungal receptors on the pDC surface. Results indicate pDCs do not phagocytose Aspergillus conidia, but instead bind hyphal surfaces and undergo activation and maturation via the upregulation of costimulatory and maturation markers. Measuring the expression of C-type lectin receptors dectin-1, dectin-2, dectin-3, and mannose receptor on human pDCs revealed intermediate expression of each receptor compared with monocytes. The specific dectin-1 agonist curdlan induced pDC activation and maturation in a cell-intrinsic and cell-extrinsic manner. The indirect activation of pDCs by curdlan was much stronger than direct stimulation and was mediated through cytokine production by other PBMCs. Overall, our data indicate pDCs express various C-type lectin receptors, recognize and respond to Aspergillus hyphal Ag, and serve as immune enhancers or modulators in the overarching fungal immune response.


Asunto(s)
Aspergillus fumigatus , Lectinas Tipo C , Humanos , Antifúngicos , Células Dendríticas , Fagocitosis
11.
Annu Rev Microbiol ; 76: 703-726, 2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-35759871

RESUMEN

Invasive fungal infections are emerging diseases that kill over 1.5 million people per year worldwide. With the increase of immunocompromised populations, the incidence of invasive fungal infections is expected to continue to rise. Vaccines for viral and bacterial infectious diseases have had a transformative impact on human health worldwide. However, no fungal vaccines are currently in clinical use. Recently, interest in fungal vaccines has grown significantly. One Candida vaccine has completed phase 2 clinical trials, and research on vaccines against coccidioidomycosis continues to advance. Additionally, multiple groups have discovered various Cryptococcus mutant strains that promote protective responses to subsequent challenge in mouse models. There has also been progress in antibody-mediated fungal vaccines. In this review, we highlight recent fungal vaccine research progress, outline the wealth of data generated, and summarize current research for both fungal biology and immunology studies relevant to fungal vaccine development. We also review technological advancements in vaccine development and highlight the future prospects of a human vaccine against invasive fungal infections.


Asunto(s)
Vacunas Fúngicas , Infecciones Fúngicas Invasoras , Vacunas , Animales , Humanos , Inmunidad , Ratones , Desarrollo de Vacunas
12.
Front Cell Infect Microbiol ; 12: 859049, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35402316

RESUMEN

Macrophages are key cellular components of innate immunity, acting as the first line of defense against pathogens to modulate homeostatic and inflammatory responses. They help clear pathogens and shape the T-cell response through the production of cytokines and chemokines. The facultative intracellular fungal pathogen Cryptococcus neoformans has developed a unique ability to interact with and manipulate host macrophages. These interactions dictate how Cryptococcus infection can remain latent or how dissemination within the host is achieved. In addition, differences in the activities of macrophages have been correlated with differential susceptibilities of hosts to Cryptococcus infection, highlighting the importance of macrophages in determining disease outcomes. There is now abundant information on the interaction between Cryptococcus and macrophages. In this review we discuss recent advances regarding macrophage origin, polarization, activation, and effector functions during Cryptococcus infection. The importance of these strategies in pathogenesis and the potential of immunotherapy for cryptococcosis treatment is also discussed.


Asunto(s)
Criptococosis , Cryptococcus neoformans , Neumonía , Criptococosis/microbiología , Cryptococcus neoformans/fisiología , Humanos , Inmunomodulación , Pulmón/patología , Macrófagos/microbiología
13.
Cell Rep ; 38(2): 110215, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35021079

RESUMEN

Macrophages are known to mediate anti-helminth responses, but it remains uncertain which subsets are involved or how macrophages actually kill helminths. Here, we show rapid monocyte recruitment to the lung after infection with the nematode parasite Nippostrongylus brasiliensis. In this inflamed tissue microenvironment, these monocytes differentiate into an alveolar macrophage (AM)-like phenotype, expressing both SiglecF and CD11c, surround invading parasitic larvae, and preferentially kill parasites in vitro. Monocyte-derived AMs (Mo-AMs) express type 2-associated markers and show a distinct remodeling of the chromatin landscape relative to tissue-derived AMs (TD-AMs). In particular, they express high amounts of arginase-1 (Arg1), which we demonstrate mediates helminth killing through L-arginine depletion. These studies indicate that recruited monocytes are selectively programmed in the pulmonary environment to express AM markers and an anti-helminth phenotype.


Asunto(s)
Pulmón/inmunología , Macrófagos Alveolares/inmunología , Infecciones por Strongylida/inmunología , Animales , Arginasa/metabolismo , Diferenciación Celular , Citocinas , Femenino , Pulmón/parasitología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Nippostrongylus , Infecciones por Strongylida/parasitología
14.
Nat Commun ; 12(1): 2624, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33976143

RESUMEN

The etiology of ulcerative colitis is poorly understood and is likely to involve perturbation of the complex interactions between the mucosal immune system and the commensal bacteria of the gut, with cytokines acting as important cross-regulators. Here we use IFN receptor-deficient mice in a dextran sulfate sodium (DSS) model of acute intestinal injury to study the contributions of type I and III interferons (IFN) to the initiation, progression and resolution of acute colitis. We find that mice lacking both types of IFN receptors exhibit enhanced barrier destruction, extensive loss of goblet cells and diminished proliferation of epithelial cells in the colon following DSS-induced damage. Impaired mucosal healing in double IFN receptor-deficient mice is driven by decreased amphiregulin expression, which IFN signaling can up-regulate in either the epithelial or hematopoietic compartment. Together, these data underscore the pleiotropic functions of IFNs and demonstrate that these critical antiviral cytokines also support epithelial regeneration following acute colonic injury.


Asunto(s)
Colitis Ulcerosa/inmunología , Interferones/metabolismo , Mucosa Intestinal/patología , Repitelización/inmunología , Animales , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/patología , Sulfato de Dextran/administración & dosificación , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Células Epiteliales , Femenino , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Masculino , Ratones , Ratones Noqueados , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Organismos Libres de Patógenos Específicos
15.
J Immunol ; 205(11): 3058-3070, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33087405

RESUMEN

RIG-I-like receptors (RLR) are cytosolic RNA sensors that signal through the MAVS adaptor to activate IFN responses against viruses. Whether the RLR family has broader effects on host immunity against other pathogen families remains to be fully explored. In this study, we demonstrate that MDA5/MAVS signaling was essential for host resistance against pulmonary Aspergillus fumigatus challenge through the regulation of antifungal leukocyte responses in mice. Activation of MDA5/MAVS signaling was driven by dsRNA from live A. fumigatus serving as a key vitality-sensing pattern recognition receptor. Interestingly, induction of type I IFNs after A. fumigatus challenge was only partially dependent on MDA5/MAVS signaling, whereas type III IFN expression was entirely dependent on MDA5/MAVS signaling. Ultimately, type I and III IFN signaling drove the expression of CXCL10. Furthermore, the MDA5/MAVS-dependent IFN response was critical for the induction of optimal antifungal neutrophil killing of A. fumigatus spores. In conclusion, our data broaden the role of the RLR family to include a role in regulating antifungal immunity against A. fumigatus.


Asunto(s)
Aspergillus fumigatus/inmunología , Helicasa Inducida por Interferón IFIH1/inmunología , Helicasa Inducida por Interferón IFIH1/metabolismo , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Quimiocina CXCL10/inmunología , Quimiocina CXCL10/metabolismo , Femenino , Interferones/inmunología , Interferones/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Reconocimiento de Patrones/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal/inmunología
16.
Artículo en Inglés | MEDLINE | ID: mdl-32733815

RESUMEN

Pulmonary infections with Aspergillus fumigatus (Af) are a significant cause of invasive fungal disease and lead to high morbidity and mortality in diverse populations throughout the world. Currently available antifungal drugs are often ineffective, thus contributing to unacceptably high mortality rates in patients suffering from invasive fungal infections. The use of cytokines as adjunctive immune therapies holds the promise of significantly improving patient outcomes in the future. In recent studies, we identified an essential role for type I and III interferons as regulators of optimal antifungal responses by pulmonary neutrophils during infection with Af. Although various membrane and cytosolic nucleic acid sensors are known to regulate interferon production in response to viruses, the pathways that regulate the production of these cytokines during fungal infection remain uncovered. In the current study, we demonstrate that dectin-1-mediated recognition of ß-glucan on the cell wall of the clinically relevant fungal pathogen Aspergillus fumigatus promotes the activation of a protective cascade of type I and III interferon expression. We further demonstrate that exogenous administration of type I and III interferons can rescue inadequate antifungal responses in dectin-1-/- mice, suggesting the potential therapeutic benefit of these cytokines as activators of antifungal defense in the context of innate defects.


Asunto(s)
Antifúngicos , Interferones , Animales , Aspergillus fumigatus , Humanos , Lectinas Tipo C , Pulmón , Ratones
17.
Cell Host Microbe ; 28(1): 104-116.e4, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32485165

RESUMEN

Aspergillus fumigatus, a ubiquitous mold, is a common cause of invasive aspergillosis (IA) in immunocompromised patients. Host defense against IA relies on lung-infiltrating neutrophils and monocyte-derived dendritic cells (Mo-DCs). Here, we demonstrate that plasmacytoid dendritic cells (pDCs), which are prototypically antiviral cells, participate in innate immune crosstalk underlying mucosal antifungal immunity. Aspergillus-infected murine Mo-DCs and neutrophils recruited pDCs to the lung by releasing the CXCR3 ligands, CXCL9 and CXCL10, in a Dectin-1 and Card9- and type I and III interferon signaling-dependent manner, respectively. During aspergillosis, circulating pDCs entered the lung in response to CXCR3-dependent signals. Via targeted pDC ablation, we found that pDCs were essential for host defense in the presence of normal neutrophil and Mo-DC numbers. Although interactions between pDC and fungal cells were not detected, pDCs regulated neutrophil NADPH oxidase activity and conidial killing. Thus, pDCs act as positive feedback amplifiers of neutrophil effector activity against inhaled mold conidia.


Asunto(s)
Aspergilosis/inmunología , Células Dendríticas/inmunología , Neutrófilos/inmunología , Receptores CXCR3/inmunología , Esporas Fúngicas/inmunología , Animales , Aspergillus fumigatus/inmunología , Proteínas Adaptadoras de Señalización CARD/inmunología , Quimiocina CXCL10/inmunología , Quimiocina CXCL9/inmunología , Inmunidad Innata , Huésped Inmunocomprometido , Interferones/inmunología , Lectinas Tipo C/inmunología , Pulmón/inmunología , Pulmón/microbiología , Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/inmunología , Receptores CCR2/inmunología , Receptores CXCR3/genética , Transducción de Señal/inmunología
18.
Semin Immunol ; 43: 101303, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31771761

RESUMEN

The unexpected discovery of a novel family of antiviral mediators, type III IFNs or IFN-λs, challenged the widely accepted primacy of type I IFNs in antiviral immunity, and it is now well recognized that the IFN-λ-based antiviral system plays a major role in antiviral protection of epithelial barriers. The recent characterization of previously unknown IFN-λ-mediated activities has prompted further reassessment of the role of type I IFNs in innate and adaptive immune and inflammatory responses. Since type I and type III IFNs are co-produced in response to a variety of stimuli, it is likely that many physiological processes are simultaneously and coordinately regulated by these cytokines in pathological conditions, and likely at steady state, as baseline expression of both IFN types is maintained by microbiota. In this review, we discuss emerging differences in the production and signaling of type I and type III IFNs, and summarize results of recent studies describing the involvement of type III IFNs in anti-bacterial and anti-fungal, as well as antiviral, defenses.


Asunto(s)
Infecciones Bacterianas/inmunología , Interferón Tipo I/metabolismo , Interferones/metabolismo , Microbiota/inmunología , Micosis/inmunología , Virosis/inmunología , Animales , Humanos , Inmunidad , Inflamación , Transducción de Señal , Interferón lambda
19.
mBio ; 10(6)2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31772051

RESUMEN

Cryptococcus neoformans is a fungal pathogen that infects the lungs and then often disseminates to the central nervous system, causing meningitis. How Cryptococcus is able to suppress host immunity and escape the antifungal activity of macrophages remains incompletely understood. We reported that the F-box protein Fbp1, a subunit of the SCF(Fbp1) E3 ligase, promotes Cryptococcus virulence by regulating host-Cryptococcus interactions. Our recent studies demonstrated that the fbp1Δ mutant elicited superior protective Th1 host immunity in the lungs and that the enhanced immunogenicity of heat-killed fbp1Δ yeast cells can be harnessed to confer protection against a subsequent infection with the virulent parental strain. We therefore examined the use of heat-killed fbp1Δ cells in several vaccination strategies. Interestingly, the vaccine protection remains effective even in mice depleted of CD4+ T cells. This finding is particularly important in the context of HIV/AIDS-induced immune deficiency. Moreover, we observed that vaccinating mice with heat-killed fbp1Δ induces significant cross-protection against challenge with diverse invasive fungal pathogens, including C. neoformans, C. gattii, and Aspergillus fumigatus, as well as partial protection against Candida albicans Thus, our data suggest that the heat-killed fbp1Δ strain has the potential to be a suitable vaccine candidate against cryptococcosis and other invasive fungal infections in both immunocompetent and immunocompromised populations.IMPORTANCE Invasive fungal infections kill more than 1.5 million people each year, with limited treatment options. There is no vaccine available in clinical use to prevent and control fungal infections. Our recent studies showed that a mutant of the F-box protein Fbp1, a subunit of the SCF(Fbp1) E3 ligase in Cryptococcus neoformans, elicited superior protective Th1 host immunity. Here, we demonstrate that the heat-killed fbp1Δ cells (HK-fbp1) can be harnessed to confer protection against a challenge by the virulent parental strain, even in animals depleted of CD4+ T cells. This finding is particularly important in the context of HIV/AIDS-induced immune deficiency. Moreover, we observed that HK-fbp1 vaccination induces significant cross-protection against challenge with diverse invasive fungal pathogens. Thus, our data suggest that HK-fbp1 has the potential to be a broad-spectrum vaccine candidate against invasive fungal infections in both immunocompetent and immunocompromised populations.


Asunto(s)
Criptococosis/prevención & control , Cryptococcus neoformans/inmunología , Vacunas Fúngicas/administración & dosificación , Infecciones Fúngicas Invasoras/prevención & control , Animales , Aspergillus fumigatus/inmunología , Aspergillus fumigatus/fisiología , Protección Cruzada , Criptococosis/inmunología , Criptococosis/microbiología , Cryptococcus gattii/inmunología , Cryptococcus gattii/fisiología , Cryptococcus neoformans/química , Cryptococcus neoformans/genética , Femenino , Proteínas Fúngicas/administración & dosificación , Proteínas Fúngicas/genética , Proteínas Fúngicas/inmunología , Vacunas Fúngicas/genética , Vacunas Fúngicas/inmunología , Calor , Humanos , Infecciones Fúngicas Invasoras/inmunología , Infecciones Fúngicas Invasoras/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/genética , Vacunas de Productos Inactivados/inmunología
20.
J Interferon Cytokine Res ; 39(10): 609-617, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31013458

RESUMEN

Neutrophils are increasingly appreciated as multifaceted regulators of innate immunity and inflammation. Historically, these important innate cells have been considered suicidal phagocytes with a primary role in the destruction of extracellular pathogens. Recent studies have significantly altered this simplistic view of neutrophils and have instead presented extensive evidence for a complex role for neutrophils in the control of diverse inflammatory conditions. It is now appreciated that neutrophils are crucial not only for efficient clearance of various pathogens but also in the development and control of inflammatory states such as autoimmunity, cancer, and tissue repair. Mounting evidence also suggests that neutrophils are capable of differential activation giving rise to distinctly polarized cells with diverse effector functions. Interferon lambda (IFN-λ) (also known as type III IFN) has emerged as an unexpected regulator of neutrophil function. IFN-λs are the newest members of the IFN family of antiviral cytokines and although initial studies suggested identical biological activities to type I IFNs, it is now apparent that type III IFN has distinct functions in vivo. In this article, I summarize recent evidence linking type III IFNs to the regulation and potential tailoring of neutrophil responses. These exciting observations might have important implications for the development of IFN-λs as novel therapeutic cytokines for the treatment of a diversity of inflammatory states where neutrophils are crucial players.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Interferones/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias/inmunología , Neutrófilos/inmunología , Animales , Enfermedades Autoinmunes/patología , Humanos , Inflamación/inmunología , Inflamación/patología , Neoplasias/patología , Neutrófilos/patología , Interferón lambda
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...