Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Br J Haematol ; 201(1): 45-57, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36484163

RESUMEN

In chronic lymphocytic leukaemia (CLL) the efficacy of SARS-CoV-2 vaccination remains unclear as most studies have focused on humoral responses. Here we comprehensively examined humoral and cellular responses to vaccine in CLL patients. Seroconversion was observed in 55.2% of CLL with lower rate and antibody titres in treated patients. T-cell responses were detected in a significant fraction of patients. CD4+ and CD8+ frequencies were significantly increased independent of serology with higher levels of CD4+ cells in patients under a Bruton tyrosine kinase (BTK) or a B-cell lymphoma 2 (BCL-2) inhibitor. Vaccination skewed CD8+ cells towards a highly cytotoxic phenotype, more pronounced in seroconverted patients. A high proportion of patients showed spike-specific CD4+ and CD8+ cells producing interferon gamma (IFNγ) and tumour necrosis factor alpha (TNFα). Patients under a BTK inhibitor showed increased production of IFNγ and TNFα by CD4+ cells. Vaccination induced a Th1 polarization reverting the Th2 CLL T-cell profile in the majority of patients with lower IL-4 production in untreated and BTK-inhibitor-treated patients. Such robust T-cell responses may have contributed to remarkable protection against hospitalization and death in a cohort of 540 patients. Combining T-cell metrics with seroprevalence may yield a more accurate measure of population immunity in CLL, providing consequential insights for public health.


Asunto(s)
Antineoplásicos , COVID-19 , Leucemia Linfocítica Crónica de Células B , Vacunas , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Vacunas contra la COVID-19/uso terapéutico , Factor de Necrosis Tumoral alfa , SARS-CoV-2 , Estudios Seroepidemiológicos , COVID-19/prevención & control , Antineoplásicos/uso terapéutico , Interferón gamma
2.
Int J Mol Sci ; 24(19)2023 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-37834316

RESUMEN

The pathogenesis of coronavirus disease 2019 (COVID-19) is associated with a hyperinflammatory response. The mechanisms of SARS-CoV-2-induced inflammation are scantly known. Methylglyoxal (MG) is a glycolysis-derived byproduct endowed with a potent glycating action, leading to the formation of advanced glycation end products (AGEs), the main one being MG-H1. MG-H1 exerts strong pro-inflammatory effects, frequently mediated by the receptor for AGEs (RAGE). Here, we investigated the involvement of the MG-H1/RAGE axis as a potential novel mechanism in SARS-CoV-2-induced inflammation by resorting to human bronchial BEAS-2B and alveolar A549 epithelial cells, expressing different levels of the ACE2 receptor (R), exposed to SARS-CoV-2 spike protein 1 (S1). Interestingly, we found in BEAS-2B cells that do not express ACE2-R that S1 exerted a pro-inflammatory action through a novel MG-H1/RAGE-based pathway. MG-H1 levels, RAGE and IL-1ß expression levels in nasopharyngeal swabs from SARS-CoV-2-positive and -negative individuals, as well as glyoxalase 1 expression, the major scavenging enzyme of MG, seem to support the results obtained in vitro. Altogether, our findings reveal a novel mechanism involved in the inflammation triggered by S1, paving the way for the study of the MG-H1/RAGE inflammatory axis in SARS-CoV-2 infection as a potential therapeutic target to mitigate COVID-19-associated pathogenic inflammation.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Glicoproteína de la Espiga del Coronavirus , Piruvaldehído/farmacología , Piruvaldehído/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Enzima Convertidora de Angiotensina 2 , Inflamación/metabolismo
3.
Infect Immun ; 90(4): e0004822, 2022 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-35311544

RESUMEN

It is becoming increasingly clear that the communities of microorganisms that populate the surfaces exposed to the external environment, termed microbiota, are key players in the regulation of pathogen-host cross talk affecting the onset as well as the outcome of infectious diseases. We have performed a multicenter, prospective, observational study in which nasal and oropharyngeal swabs were collected for microbiota predicting the risk of invasive fungal infections (IFIs) in patients with hematological malignancies. Here, we demonstrate that the nasal and oropharyngeal microbiota are different, although similar characteristics differentiate high-risk from low-risk samples at both sites. Indeed, similar to previously published results on the oropharyngeal microbiota, high-risk samples in the nose were characterized by low diversity, a loss of beneficial bacteria, and an expansion of potentially pathogenic taxa, in the presence of reduced levels of tryptophan (Trp). At variance with oropharyngeal samples, however, low Trp levels were associated with defective host-derived kynurenine production, suggesting reduced tolerance mechanisms at the nasal mucosal surface. This was accompanied by reduced levels of the chemokine interleukin-8 (IL-8), likely associated with a reduced recruitment of neutrophils and impaired fungal clearance. Thus, the nasal and pharyngeal microbiomes of hematological patients provide complementary information that could improve predictive tools for the risk of IFI in hematological patients.


Asunto(s)
Infecciones Fúngicas Invasoras , Microbiota , Bacterias , Humanos , Nariz/microbiología , Estudios Prospectivos
4.
Eur J Clin Microbiol Infect Dis ; 40(1): 1-12, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33037944

RESUMEN

Can a patient diagnosed with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) be infected again? This question is still unsolved. We tried to analyze local and literature cases with a positive respiratory swab after recovery. We collected data from symptomatic patients diagnosed with SARS-CoV-2 infection in the Italian Umbria Region that, after recovery, were again positive for SARS-CoV-2 in respiratory tract specimens. Samples were also assessed for infectivity in vitro. A systematic review of similar cases reported in the literature was performed. The study population was composed of 9 patients during a 4-month study period. Among the new positive samples, six were inoculated in Vero-E6 cells and showed no growth and negative molecular test in culture supernatants. All patients were positive for IgG against SARS-CoV-2 nucleoprotein and/or S protein. Conducting a review of the literature, 1350 similar cases have been found. The presumptive reactivation occurred in 34.5 days on average (standard deviation, SD, 18.7 days) after COVID-19 onset, when the 5.6% of patients presented fever and the 27.6% symptoms. The outcome was favorable in 96.7% of patients, while the 1.1% of them were still hospitalized at the time of data collection and the 2.1% died. Several hypotheses have been formulated to explain new positive respiratory samples after confirmed negativity. According to this study, the phenomenon seems to be due to the prolonged detection of SARS-CoV-2 RNA traces in respiratory samples of recovered patients. The failure of the virus to replicate in vitro suggests its inability to replicate in vivo.


Asunto(s)
Prueba de COVID-19/estadística & datos numéricos , COVID-19/diagnóstico , COVID-19/fisiopatología , Adulto , Anciano , Animales , Chlorocebus aethiops , Femenino , Humanos , Italia , Masculino , Persona de Mediana Edad , Nasofaringe/virología , ARN Viral/análisis , Recurrencia , Células Vero , Replicación Viral
5.
Eur J Immunol ; 49(1): 195-198, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30267564

RESUMEN

Intravenous immunoglobuin (IVIG) exerts protective effects in experimental allergic bronchopulmonary aspergillosis (ABPA) via a sialylation-dependent mechanism. The protection was associated with reduced recruitment of eosinophils, diminished goblet cell hyperplasia, suppressed Th2 and Th17 responses and reciprocally enhanced regulatory T cells and IL-10, and decreased IgE levels in the circulation.


Asunto(s)
Aspergilosis Broncopulmonar Alérgica/terapia , Eosinófilos/inmunología , Células Caliciformes/inmunología , Inmunoglobulinas Intravenosas/uso terapéutico , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Células Th2/inmunología , Animales , Células Cultivadas , Humanos , Inmunoglobulina E/sangre , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos C57BL , Ácido N-Acetilneuramínico/metabolismo
6.
Clin Exp Rheumatol ; 38(4): 754-759, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32723434

RESUMEN

OBJECTIVES: COVID-19 features include disseminated intravascular coagulation and thrombotic microangiopathy indicating a hypercoagulable state. We aimed to investigate antiphospholipid antibodies (aPL) prevalence and clinical relationships in a large cohort of COVID-19 patients. METHODS: We analysed the prevalence and titres of serum aPL in 122 patients with COVID-19 and 157 with primary antiphospholipid syndrome (PAPS) and 91 with other autoimmune rheumatic diseases (oARD) for comparison. IgG/IgM anticardiolipin (aCL) and IgG/IgM anti-beta2glycoprotein I (ß2GPI) were assayed using homemade ELISA, IgA aCL and anti-ß2GPI by commercial ELISA kits and lupus anticoagulant (LAC) by multiple coagulation tests following updated international guidelines. RESULTS: Prevalence of IgG and IgM aCL and of IgG and IgM anti-ß2GPI across COVID-19 patients were 13.4%, 2.7%, 6.3% and 7.1%, being significantly lower than in PAPS (p<0.0001 for all). Frequency of IgG aCL and IgM anti-ß2GPI was comparable to oARD (13.4% vs. 13.2% and 7.1% vs. 11%, respectively), while IgG anti-ß2GPI and IgM aCL were lower (p<0.01). IgA aCL and IgA anti-ß2GPI were retrieved in 1.7% and 3.3% of COVID-19 patients, respectively. Positive LAC was observed in 22.2% COVID-19 vs. 54.1% of PAPS (p<0.0001) and 14.6% of oARD (p=0.21). Venous or arterial thromboses occurred in 18/46 (39.1%) COVID-19 patients and were not associated with positive aPL (p=0.09). CONCLUSIONS: Thrombosis is a frequent manifestation during COVID-19 infection. However, prevalence and titres of aPL antibodies or LAC were neither consistently increased nor associated with thrombosis when measured at a single timepoint, therefore not representing a suitable screening tool in the acute stage of disease.


Asunto(s)
Anticuerpos Antifosfolípidos/sangre , Infecciones por Coronavirus/inmunología , Neumonía Viral/inmunología , Anticuerpos Anticardiolipina/sangre , Síndrome Antifosfolípido/sangre , Betacoronavirus , COVID-19 , Infecciones por Coronavirus/sangre , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Pandemias , Neumonía Viral/sangre , SARS-CoV-2 , Trombosis/complicaciones , Trombosis/virología , beta 2 Glicoproteína I/inmunología
7.
Blood ; 125(20): 3173-82, 2015 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-25769621

RESUMEN

Because activating killer cell immunoglobulinlike receptors (KIRs) are heterogeneously expressed in the population, we investigated the role of donor activating KIRs in haploidentical hematopoietic transplants for acute leukemia. Transplants were grouped according to presence vs absence of KIR-ligand mismatches in the graft-vs-host direction (ie, of donor-vs-recipient natural killer [NK]-cell alloreactivity). In the absence of donor-vs-recipient NK-cell alloreactivity, donor activating KIRs had no effects on outcomes. In the 69 transplant pairs with donor-vs-recipient NK-cell alloreactivity, transplantation from donors with KIR2DS1 and/or KIR3DS1 was associated with reduced risk of nonrelapse mortality, largely infection related (KIR2DS1 present vs absent: hazard ratio [HR], 0.25; P = .01; KIR3DS1 present vs absent: HR, 0.18; P = .006), and better event-free survival (KIR2DS1 present vs absent: HR, 0.31; P = .011; KIR3DS1 present vs absent: HR, 0.30; P = .008). Transplantation from donors with KIR2DS1 and/or KIR3DS1 was also associated with a 50% reduction in infection rate (P = .003). In vitro analyses showed that KIR2DS1 binding to its HLA-C2 ligand upregulated inflammatory cytokine production by alloreactive NK cells in response to infectious challenges. Because ∼40% of donors able to exert donor-vs-recipient NK-cell alloreactivity carry KIR2DS1 and/or KIR3DS1, searching for them may become a feasible, additional criterion in donor selection.


Asunto(s)
Haplotipos , Trasplante de Células Madre Hematopoyéticas , Receptores KIR/genética , Receptores KIR/metabolismo , Donantes de Tejidos , Sitios Genéticos , Genotipo , Enfermedad Injerto contra Huésped/etiología , Antígenos HLA-C/inmunología , Antígenos HLA-C/metabolismo , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Leucemia/genética , Leucemia/mortalidad , Leucemia/patología , Leucemia/terapia , Estadificación de Neoplasias , Unión Proteica , Estudios Retrospectivos , Trasplante Homólogo
8.
PLoS Pathog ; 10(11): e1004462, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25375146

RESUMEN

Since IL-37 transgenic mice possesses broad anti-inflammatory properties, we assessed whether recombinant IL-37 affects inflammation in a murine model of invasive pulmonary aspergillosis. Recombinant human IL-37 was injected intraperitoneally into mice prior to infection and the effects on lung inflammation and inflammasome activation were evaluated. IL-37 markedly reduced NLRP3-dependent neutrophil recruitment and steady state mRNA levels of IL-1ß production and mitigated lung inflammation and damage in a relevant clinical model, namely aspergillosis in mice with cystic fibrosis. The anti-inflammatory activity of IL-37 requires the IL-1 family decoy receptor TIR-8/SIGIRR. Thus, by preventing activation of the NLRP3 inflammasome and reducing IL-1ß secretion, IL-37 functions as a broad spectrum inhibitor of the innate response to infection-mediated inflammation, and could be considered to be therapeutic in reducing the pulmonary damage due to non-resolving Aspergillus infection and disease.


Asunto(s)
Inflamasomas/inmunología , Interleucina-1/inmunología , Aspergilosis Pulmonar/inmunología , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Modelos Animales de Enfermedad , Femenino , Inflamasomas/genética , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-1/genética , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Aspergilosis Pulmonar/genética , Aspergilosis Pulmonar/patología , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/inmunología
9.
PLoS Pathog ; 10(3): e1003936, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24603878

RESUMEN

The galactosaminogalactan (GAG) is a cell wall component of Aspergillus fumigatus that has potent anti-inflammatory effects in mice. However, the mechanisms responsible for the anti-inflammatory property of GAG remain to be elucidated. In the present study we used in vitro PBMC stimulation assays to demonstrate, that GAG inhibits proinflammatory T-helper (Th)1 and Th17 cytokine production in human PBMCs by inducing Interleukin-1 receptor antagonist (IL-1Ra), a potent anti-inflammatory cytokine that blocks IL-1 signalling. GAG cannot suppress human T-helper cytokine production in the presence of neutralizing antibodies against IL-1Ra. In a mouse model of invasive aspergillosis, GAG induces IL-1Ra in vivo, and the increased susceptibility to invasive aspergillosis in the presence of GAG in wild type mice is not observed in mice deficient for IL-1Ra. Additionally, we demonstrate that the capacity of GAG to induce IL-1Ra could also be used for treatment of inflammatory diseases, as GAG was able to reduce severity of an experimental model of allergic aspergillosis, and in a murine DSS-induced colitis model. In the setting of invasive aspergillosis, GAG has a significant immunomodulatory function by inducing IL-1Ra and notably IL-1Ra knockout mice are completely protected to invasive pulmonary aspergillosis. This opens new treatment strategies that target IL-1Ra in the setting of acute invasive fungal infection. However, the observation that GAG can also protect mice from allergy and colitis makes GAG or a derivative structure of GAG a potential treatment compound for IL-1 driven inflammatory diseases.


Asunto(s)
Aspergilosis/inmunología , Aspergillus fumigatus/inmunología , Polisacáridos Fúngicos/inmunología , Proteína Antagonista del Receptor de Interleucina 1/biosíntesis , Polisacáridos/inmunología , Factores de Virulencia/inmunología , Animales , Citocinas/biosíntesis , Citocinas/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Proteína Antagonista del Receptor de Interleucina 1/inmunología , Leucocitos Mononucleares/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados
10.
J Immunol ; 193(5): 2340-8, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25049357

RESUMEN

The long pentraxin 3 (PTX3) modulates different effector pathways involved in innate resistance to Aspergillus fumigatus, including complement activation or promotion of phagocytosis by interacting with FcγRs. However, whether and how TLRs modulate PTX3 mediates antifungal resistance is not known. In this study, we demonstrate that PTX3 binds myeloid differentiation protein 2 (MD-2) in vitro and exerts its protective antifungal activity in vivo through TLR4/MD-2-mediated signaling. Similar to Tlr4(-/-) mice, Md2(-/-) mice displayed high susceptibility to pulmonary aspergillosis, a phenotype associated with a proinflammatory cytokine profile and impaired antifungal activity of polymorphonuclear neutrophils. Treating Md2(-/-) mice with PTX3 failed to confer immune protection against the fungus, whereas adoptive transfer of MD-2-competent polymorphonuclear neutrophils restored it. Mechanistically, engagement of MD-2 by PTX3-opsonized Aspergillus conidia activated the TLR4/Toll/IL-1R domain-containing adapter inducing IFN-ß-dependent signaling pathway converging on IL-10. Thus, we have identified a novel receptor mechanism, involving the TLR4/MD-2/Toll/IL-1R domain-containing adapter inducing IFN-ß-mediated signaling, whereby PTX3 elicits antifungal resistance with limited immunopathology in A. fumigatus infection.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/inmunología , Aspergilosis/inmunología , Aspergillus fumigatus/inmunología , Proteína C-Reactiva/inmunología , Antígeno 96 de los Linfocitos/inmunología , Proteínas del Tejido Nervioso/inmunología , Componente Amiloide P Sérico/inmunología , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Aspergilosis/genética , Aspergilosis/patología , Aspergillus fumigatus/genética , Proteína C-Reactiva/genética , Células HEK293 , Humanos , Interferón beta/genética , Interferón beta/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Antígeno 96 de los Linfocitos/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Componente Amiloide P Sérico/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
11.
Proc Natl Acad Sci U S A ; 110(27): 11079-84, 2013 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-23776239

RESUMEN

Many fungi restructured their proteomes through incorporation of serine (Ser) at thousands of protein sites coded by the leucine (Leu) CUG codon. How these fungi survived this potentially lethal genetic code alteration and its relevance for their biology are not understood. Interestingly, the human pathogen Candida albicans maintains variable Ser and Leu incorporation levels at CUG sites, suggesting that this atypical codon assignment flexibility provided an effective mechanism to alter the genetic code. To test this hypothesis, we have engineered C. albicans strains to misincorporate increasing levels of Leu at protein CUG sites. Tolerance to the misincorporations was very high, and one strain accommodated the complete reversion of CUG identity from Ser back to Leu. Increasing levels of Leu misincorporation decreased growth rate, but production of phenotypic diversity on a phenotypic array probing various metabolic networks, drug resistance, and host immune cell responses was impressive. Genome resequencing revealed an increasing number of genotype changes at polymorphic sites compared with the control strain, and 80% of Leu misincorporation resulted in complete loss of heterozygosity in a large region of chromosome V. The data unveil unanticipated links between gene translational fidelity, proteome instability and variability, genome diversification, and adaptive phenotypic diversity. They also explain the high heterozygosity of the C. albicans genome and open the door to produce microorganisms with genetic code alterations for basic and applied research.


Asunto(s)
Candida albicans/genética , Código Genético , Genoma Fúngico , Inestabilidad Genómica , Proteoma/genética , Animales , Candida albicans/química , Candida albicans/patogenicidad , Codón/genética , Células Dendríticas/química , Células Dendríticas/metabolismo , Evolución Molecular , Femenino , Proteínas Fúngicas/genética , Tamización de Portadores Genéticos , Variación Genética , Humanos , Ratones , Ratones Endogámicos C57BL , Fenotipo , Polimorfismo de Nucleótido Simple , ARN de Hongos/genética
12.
PLoS Pathog ; 9(11): e1003716, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24244155

RESUMEN

α-(1,3)-Glucan is a major component of the cell wall of Aspergillus fumigatus, an opportunistic human fungal pathogen. There are three genes (AGS1, AGS2 and AGS3) controlling the biosynthesis of α-(1,3)-glucan in this fungal species. Deletion of all the three AGS genes resulted in a triple mutant that was devoid of α-(1,3)-glucan in its cell wall; however, its growth and germination was identical to that of the parental strain in vitro. In the experimental murine aspergillosis model, this mutant was less pathogenic than the parental strain. The AGS deletion resulted in an extensive structural modification of the conidial cell wall, especially conidial surface where the rodlet layer was covered by an amorphous glycoprotein matrix. This surface modification was responsible for viability reduction of conidia in vivo, which explains decrease in the virulence of triple agsΔ mutant.


Asunto(s)
Aspergilosis/enzimología , Aspergillus fumigatus/enzimología , Aspergillus fumigatus/patogenicidad , Pared Celular/enzimología , Polisacáridos Fúngicos/biosíntesis , Proteínas Fúngicas/metabolismo , Glucosiltransferasas/metabolismo , Animales , Aspergilosis/genética , Aspergilosis/patología , Aspergillus fumigatus/genética , Pared Celular/genética , Polisacáridos Fúngicos/genética , Proteínas Fúngicas/genética , Eliminación de Gen , Glucosiltransferasas/genética , Humanos , Ratones Noqueados , Esporas Fúngicas/enzimología , Esporas Fúngicas/genética
13.
Nature ; 460(7259): 1117-21, 2009 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-19713928

RESUMEN

The air we breathe is filled with thousands of fungal spores (conidia) per cubic metre, which in certain composting environments can easily exceed 10(9) per cubic metre. They originate from more than a hundred fungal species belonging mainly to the genera Cladosporium, Penicillium, Alternaria and Aspergillus. Although these conidia contain many antigens and allergens, it is not known why airborne fungal microflora do not activate the host innate immune cells continuously and do not induce detrimental inflammatory responses following their inhalation. Here we show that the surface layer on the dormant conidia masks their recognition by the immune system and hence prevents immune response. To explore this, we used several fungal members of the airborne microflora, including the human opportunistic fungal pathogen Aspergillus fumigatus, in in vitro assays with dendritic cells and alveolar macrophages and in in vivo murine experiments. In A. fumigatus, this surface 'rodlet layer' is composed of hydrophobic RodA protein covalently bound to the conidial cell wall through glycosylphosphatidylinositol-remnants. RodA extracted from conidia of A. fumigatus was immunologically inert and did not induce dendritic cell or alveolar macrophage maturation and activation, and failed to activate helper T-cell immune responses in vivo. The removal of this surface 'rodlet/hydrophobin layer' either chemically (using hydrofluoric acid), genetically (DeltarodA mutant) or biologically (germination) resulted in conidial morphotypes inducing immune activation. All these observations show that the hydrophobic rodlet layer on the conidial cell surface immunologically silences airborne moulds.


Asunto(s)
Antígenos Fúngicos/inmunología , Esporas Fúngicas/inmunología , Traslado Adoptivo , Microbiología del Aire , Alérgenos , Animales , Antígenos Fúngicos/química , Antígenos Fúngicos/genética , Antígenos de Plantas , Aspergillus fumigatus/química , Aspergillus fumigatus/inmunología , Aspergillus fumigatus/fisiología , Linfocitos T CD4-Positivos/inmunología , Catepsinas , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Proteínas Fúngicas , Humanos , Ácido Fluorhídrico/química , Sistema Inmunológico/inmunología , Activación de Linfocitos , Macrófagos Alveolares/inmunología , Ratones , Ratones Endogámicos C57BL , Esporas Fúngicas/química , Esporas Fúngicas/genética
14.
J Antimicrob Chemother ; 69(4): 1065-74, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24265229

RESUMEN

OBJECTIVES: Micafungin inhibits 1,3-ß-D-glucan synthase and interferes with fungal cell wall synthesis. Clinically, micafungin has been shown to be efficacious for the treatment of invasive fungal infections. However, little is known about the immunomodulatory activity of micafungin in these infections. METHODS: We evaluated the immunomodulatory activity of escalating doses of micafungin in murine and human polymorphonuclear neutrophils (PMNs) in vitro and in vivo in different preclinical models of invasive aspergillosis, including mice deficient for selected innate immune receptors. RESULTS: Micafungin was able to regulate PMN cytokine response to Aspergillus fumigatus conidia by decreasing the expression of tumour necrosis factor-α and increasing that of interleukin-10 (IL-10). In vivo, the therapeutic efficacy of micafungin was strictly dose-dependent, with the maximum activity observed at the highest dose, concomitant with reduced inflammatory pathology. The anti-inflammatory activity of micafungin required IL-10 and occurred through signalling via the TLR2/dectin-1 and TLR3/TRIF pathways. CONCLUSION: Together, these findings suggest that the therapeutic efficacy of micafungin in aspergillosis is orchestrated by the activation of innate immune receptors affecting the inflammatory/anti-inflammatory balance during infection.


Asunto(s)
Aspergilosis/tratamiento farmacológico , Aspergillus fumigatus/efectos de los fármacos , Equinocandinas/uso terapéutico , Factores Inmunológicos/uso terapéutico , Lipopéptidos/uso terapéutico , Animales , Aspergilosis/microbiología , Carga Bacteriana , Células Cultivadas , Equinocandinas/farmacología , Femenino , Histocitoquímica , Humanos , Factores Inmunológicos/farmacología , Lipopéptidos/farmacología , Pulmón/microbiología , Pulmón/patología , Micafungina , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neutrófilos/efectos de los fármacos , Neutrófilos/microbiología , Resultado del Tratamiento
15.
Blood ; 119(4): 967-77, 2012 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-22147891

RESUMEN

Aspergillus fumigatus is a model fungal pathogen and a common cause of severe infections and diseases. CD8⁺ T cells are present in the human and murine T-cell repertoire to the fungus. However, CD8⁺ T-cell function in infection and the molecular mechanisms that control their priming and differentiation into effector and memory cells in vivo remain elusive. In the present study, we report that both CD4⁺ and CD8⁺ T cells mediate protective memory responses to the fungus contingent on the nature of the fungal vaccine. Mechanistically, class I MHC-restricted, CD8⁺ memory T cells were activated through TLR3 sensing of fungal RNA by cross-presenting dendritic cells. Genetic deficiency of TLR3 was associated with susceptibility to aspergillosis and concomitant failure to activate memory-protective CD8⁺ T cells both in mice and in patients receiving stem-cell transplantations. Therefore, TLR3 essentially promotes antifungal memory CD8⁺ T-cell responses and its deficiency is a novel susceptibility factor for aspergillosis in high-risk patients.


Asunto(s)
Aspergilosis/inmunología , Aspergillus fumigatus/inmunología , Linfocitos T CD8-positivos/inmunología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Huésped Inmunocomprometido , Memoria Inmunológica , Receptor Toll-Like 3/metabolismo , Animales , Presentación de Antígeno , Antígenos Fúngicos/uso terapéutico , Aspergilosis/genética , Aspergilosis/prevención & control , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Estudios de Cohortes , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Susceptibilidad a Enfermedades , Femenino , Vacunas Fúngicas/uso terapéutico , Humanos , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Polimorfismo de Nucleótido Simple , Organismos Libres de Patógenos Específicos , Receptor Toll-Like 3/genética
16.
Nat Med ; 13(5): 579-86, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17417651

RESUMEN

Glucocorticoid-induced tumor necrosis factor receptor (GITR) on T cells and its natural ligand, GITRL, on accessory cells contribute to the control of immune homeostasis. Here we show that reverse signaling through GITRL after engagement by soluble GITR initiates the immunoregulatory pathway of tryptophan catabolism in mouse plasmacytoid dendritic cells, by means of noncanonical NF-kappaB-dependent induction of indoleamine 2,3-dioxygenase (IDO). The synthetic glucocorticoid dexamethasone administered in vivo activated IDO through the symmetric induction of GITR in CD4(+) T cells and GITRL in plasmacytoid dendritic cells. The drug exerted IDO-dependent protection in a model of allergic airway inflammation. Modulation of tryptophan catabolism via the GITR-GITRL coreceptor system might represent an effective therapeutic target in immune regulation. Induction of IDO could be an important mechanism underlying the anti-inflammatory action of corticosteroids.


Asunto(s)
Dexametasona/farmacología , Hipersensibilidad/prevención & control , Hipersensibilidad/fisiopatología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Receptores de Factor de Crecimiento Nervioso/fisiología , Receptores del Factor de Necrosis Tumoral/fisiología , Transducción de Señal/fisiología , Animales , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Proteína Relacionada con TNFR Inducida por Glucocorticoide , Humanos , Ratones , Receptores de Factor de Crecimiento Nervioso/efectos de los fármacos , Receptores del Factor de Necrosis Tumoral/efectos de los fármacos , Bazo/inmunología , Factores de Necrosis Tumoral/fisiología
17.
Front Biosci (Landmark Ed) ; 29(2): 59, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38420819

RESUMEN

BACKGROUND: The recent Coronavirus Disease 2019 (COVID-19) pandemic has dramatically exposed our gap in understanding the pathogenesis of airborne infections. Within such a context, it is increasingly clear that the nasal cavity represents a critical checkpoint not only in the initial colonization phase but also in shaping any infectious sequelae. This is particularly relevant to COVID-19 in that the nasal cavity is characterized by high-level expression of the Severe Acute Respiratory Syndrome CoronaVirus 2 (SARS-CoV-2) receptor, Angiotensin-Converting Enzyme 2 (ACE2), all along the respiratory tract. As part of the nasal mucosa, commensal microbes harbored by the nasal cavity likely are far more than just innocent bystanders in the interaction between SARS-CoV-2 and the local microenvironment. Yet the role of the qualitative composition of the nasal microbiome is unclear, as is its function, whether protective or not. METHODS: In this study, individuals undergoing SARS-CoV-2 molecular testing at the Hospital of Perugia (Italy) were recruited, with their residual material from the nasopharyngeal swabs being collected for microbiome composition analysis and short-chain fatty acid (SCFA) measurements (by 16S rRNA sequencing and gas chromatography-mass spectrometry), respectively. RESULTS: After stratification by age, gender, and viral load, the composition of the nasopharyngeal microbiome appeared to be influenced by age and gender, and SARS-CoV-2 infection further determined compositional changes. Notwithstanding this variability, a restricted analysis of female subjects-once SARS-CoV-2-infected-unraveled a shared expansion of Lachnospirales-Lachnospiraceae, irrespective of the viral load and age. This was associated with a reduction in the branched SCFA isobutanoic acid, as well as in the SCFAs with longer chains. CONCLUSIONS: Our results indicate that the nasopharyngeal microbiome is influenced by age, gender, and viral load, with consistent patterns of microbiome changes being present across specific groups. This may help in designing a personalized medicine approach in COVID-19 patients with specific patterns of nasal microbial communities.


Asunto(s)
COVID-19 , Microbiota , Humanos , Femenino , SARS-CoV-2 , ARN Ribosómico 16S/genética , Nasofaringe
18.
J Exp Med ; 204(4): 793-804, 2007 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-17389238

RESUMEN

The long pentraxin (PTX) 3 is produced by macrophages and myeloid dendritic cells in response to Toll-like receptor agonists and represents a nonredundant component of humoral innate immunity against selected pathogens. We report that, unexpectedly, PTX3 is stored in specific granules and undergoes release in response to microbial recognition and inflammatory signals. Released PTX3 can partially localize in neutrophil extracellular traps formed by extruded DNA. Eosinophils and basophils do not contain preformed PTX3. PTX3-deficient neutrophils have defective microbial recognition and phagocytosis, and PTX3 is nonredundant for neutrophil-mediated resistance against Aspergillus fumigatus. Thus, neutrophils serve as a reservoir, ready for rapid release, of the long PTX3, a key component of humoral innate immunity with opsonic activity.


Asunto(s)
Proteína C-Reactiva/metabolismo , Gránulos Citoplasmáticos/metabolismo , Espacio Extracelular/metabolismo , Neutrófilos/citología , Componente Amiloide P Sérico/metabolismo , Animales , Proteína C-Reactiva/deficiencia , Proteína C-Reactiva/genética , Células Cultivadas , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/metabolismo , Unión Proteica , Componente Amiloide P Sérico/deficiencia , Componente Amiloide P Sérico/genética
19.
PLoS Pathog ; 7(11): e1002372, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22102815

RESUMEN

A new polysaccharide secreted by the human opportunistic fungal pathogen Aspergillus fumigatus has been characterized. Carbohydrate analysis using specific chemical degradations, mass spectrometry, ¹H and ¹³C nuclear magnetic resonance showed that this polysaccharide is a linear heterogeneous galactosaminogalactan composed of α1-4 linked galactose and α1-4 linked N-acetylgalactosamine residues where both monosacharides are randomly distributed and where the percentage of galactose per chain varied from 15 to 60%. This polysaccharide is antigenic and is recognized by a majority of the human population irrespectively of the occurrence of an Aspergillus infection. GalNAc oligosaccharides are an essential epitope of the galactosaminogalactan that explains the universal antibody reaction due to cross reactivity with other antigenic molecules containing GalNAc stretches such as the N-glycans of Campylobacter jejuni. The galactosaminogalactan has no protective effect during Aspergillus infections. Most importantly, the polysaccharide promotes fungal development in immunocompetent mice due to its immunosuppressive activity associated with disminished neutrophil infiltrates.


Asunto(s)
Antígenos Fúngicos/inmunología , Aspergilosis/inmunología , Aspergillus fumigatus/inmunología , Inmunosupresores , Polisacáridos/química , Polisacáridos/inmunología , Animales , Anticuerpos Antifúngicos/inmunología , Apoptosis , Aspergillus fumigatus/metabolismo , Conformación de Carbohidratos , Secuencia de Carbohidratos , Pared Celular/inmunología , Reacciones Cruzadas , Epítopos , Femenino , Interacciones Huésped-Patógeno , Humanos , Macrófagos/inmunología , Espectroscopía de Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila , Neutrófilos/inmunología , Neutrófilos/fisiología , Polisacáridos/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
20.
Blood ; 117(22): 5881-91, 2011 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-21441461

RESUMEN

T cell-mediated heterologous immunity to different pathogens is promising for the development of immunotherapeutic strategies. Aspergillus fumigatus and Candida albicans, the 2 most common fungal pathogens causing severe infections in immunocompromised patients, are controlled by CD4+ type 1 helper T (T(H)1) cells in humans and mice, making induction of fungus-specific CD4+ T(H)1 immunity an appealing strategy for antifungal therapy. We identified an immunogenic epitope of the A fumigatus cell wall glucanase Crf1 that can be presented by 3 common major histocompatibility complex class II alleles and that induces memory CD4+ T(H)1 cells with a diverse T-cell receptor repertoire that is cross-reactive to C albicans. In BALB/c mice, the Crf1 protein also elicits cross-protection against lethal infection with C albicans that is mediated by the same epitope as in humans. These data illustrate the existence of T cell-based cross-protection for the 2 distantly related clinically relevant fungal pathogens that may foster the development of immunotherapeutic strategies.


Asunto(s)
Aspergillus fumigatus/inmunología , Candida albicans/inmunología , Candidiasis/inmunología , Candidiasis/prevención & control , Protección Cruzada , Células TH1/inmunología , Animales , Aspergillus fumigatus/patogenicidad , Western Blotting , Candida albicans/patogenicidad , Proliferación Celular , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Glicósido Hidrolasas/inmunología , Humanos , Inmunidad Celular , Interferón gamma/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fragmentos de Péptidos/inmunología , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología , Vacunación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA