Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Immunol ; 24(11): 1879-1889, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37872315

RESUMO

Gastrointestinal fungal dysbiosis is a hallmark of several diseases marked by systemic immune activation. Whether persistent pathobiont colonization during immune alterations and impaired gut barrier function has a durable impact on host immunity is unknown. We found that elevated levels of Candida albicans immunoglobulin G (IgG) antibodies marked patients with severe COVID-19 (sCOVID-19) who had intestinal Candida overgrowth, mycobiota dysbiosis and systemic neutrophilia. Analysis of hematopoietic stem cell progenitors in sCOVID-19 revealed transcriptional changes in antifungal immunity pathways and reprogramming of granulocyte myeloid progenitors (GMPs) for up to a year. Mice colonized with C. albicans patient isolates experienced increased lung neutrophilia and pulmonary NETosis during severe acute respiratory syndrome coronavirus-2 infection, which were partially resolved with antifungal treatment or by interleukin-6 receptor blockade. sCOVID-19 patients treated with tocilizumab experienced sustained reductions in C. albicans IgG antibodies titers and GMP transcriptional changes. These findings suggest that gut fungal pathobionts may contribute to immune activation during inflammatory diseases, offering potential mycobiota-immune therapeutic strategies for sCOVID-19 with prolonged symptoms.


Assuntos
COVID-19 , Micobioma , Humanos , Animais , Camundongos , Antifúngicos , Disbiose , Neutrófilos , Candida albicans , Imunoglobulina G
2.
Semin Immunol ; 67: 101757, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37003056

RESUMO

The dynamic and complex community of microbes that colonizes the intestines is composed of bacteria, fungi, and viruses. At the mucosal surfaces, immunoglobulins play a key role in protection against bacterial and fungal pathogens, and their toxins. Secretory immunoglobulin A (sIgA) is the most abundantly produced antibody at the mucosal surfaces, while Immunoglobulin G (IgG) isotypes play a critical role in systemic protection. IgA and IgG antibodies with reactivity to commensal fungi play an important role in shaping the mycobiota and host antifungal immunity. In this article, we review the latest evidence that establishes a connection between commensal fungi and B cell-mediated antifungal immunity as an additional layer of protection against fungal infections and inflammation.


Assuntos
Antifúngicos , Imunoglobulina A Secretora , Humanos , Imunoglobulina G , Bactérias , Imunidade nas Mucosas , Imunoglobulinas
4.
Nature ; 603(7902): 672-678, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35296857

RESUMO

The fungal microbiota (mycobiota) is an integral part of the complex multikingdom microbial community colonizing the mammalian gastrointestinal tract and has an important role in immune regulation1-6. Although aberrant changes in the mycobiota have been linked to several diseases, including inflammatory bowel disease3-9, it is currently unknown whether fungal species captured by deep sequencing represent living organisms and whether specific fungi have functional consequences for disease development in affected individuals. Here we developed a translational platform for the functional analysis of the mycobiome at the fungal-strain- and patient-specific level. Combining high-resolution mycobiota sequencing, fungal culturomics and genomics, a CRISPR-Cas9-based fungal strain editing system, in vitro functional immunoreactivity assays and in vivo models, this platform enables the examination of host-fungal crosstalk in the human gut. We discovered a rich genetic diversity of opportunistic Candida albicans strains that dominate the colonic mucosa of patients with inflammatory bowel disease. Among these human-gut-derived isolates, strains with high immune-cell-damaging capacity (HD strains) reflect the disease features of individual patients with ulcerative colitis and aggravated intestinal inflammation in vivo through IL-1ß-dependent mechanisms. Niche-specific inflammatory immunity and interleukin-17A-producing T helper cell (TH17 cell) antifungal responses by HD strains in the gut were dependent on the C. albicans-secreted peptide toxin candidalysin during the transition from a benign commensal to a pathobiont state. These findings reveal the strain-specific nature of host-fungal interactions in the human gut and highlight new diagnostic and therapeutic targets for diseases of inflammatory origin.


Assuntos
Fungos , Microbioma Gastrointestinal , Doenças Inflamatórias Intestinais , Microbiota , Micobioma , Animais , Sistemas CRISPR-Cas , Candida albicans , Fungos/genética , Fungos/patogenicidade , Variação Genética , Humanos , Imunidade , Inflamação , Mamíferos
5.
Nat Microbiol ; 6(12): 1493-1504, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34811531

RESUMO

Secretory immunoglobulin A (sIgA) plays an important role in gut barrier protection by shaping the resident microbiota community, restricting the growth of bacterial pathogens and enhancing host protective immunity via immunological exclusion. Here, we found that a portion of the microbiota-driven sIgA response is induced by and directed towards intestinal fungi. Analysis of the human gut mycobiota bound by sIgA revealed a preference for hyphae, a fungal morphotype associated with virulence. Candida albicans was a potent inducer of IgA class-switch recombination among plasma cells, via an interaction dependent on intestinal phagocytes and hyphal programming. Characterization of sIgA affinity and polyreactivity showed that hyphae-associated virulence factors were bound by these antibodies and that sIgA influenced C. albicans morphotypes in the murine gut. Furthermore, an increase in granular hyphal morphologies in patients with Crohn's disease compared with healthy controls correlated with a decrease in antifungal sIgA antibody titre with affinity to two hyphae-associated virulence factors. Thus, in addition to its importance in gut bacterial regulation, sIgA targets the uniquely fungal phenomenon of hyphal formation. Our findings indicate that antifungal sIgA produced in the gut can play a role in regulating intestinal fungal commensalism by coating fungal morphotypes linked to virulence, thereby providing a protective mechanism that might be dysregulated in patients with Crohn's disease.


Assuntos
Doença de Crohn/microbiologia , Fungos/fisiologia , Microbioma Gastrointestinal , Imunoglobulina A Secretora/imunologia , Simbiose , Animais , Candida albicans/genética , Candida albicans/fisiologia , Doença de Crohn/genética , Doença de Crohn/imunologia , Feminino , Fungos/genética , Interações Hospedeiro-Patógeno , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fagócitos/imunologia , Fagócitos/microbiologia
6.
Cell ; 184(4): 1017-1031.e14, 2021 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-33548172

RESUMO

Antibodies mediate natural and vaccine-induced immunity against viral and bacterial pathogens, whereas fungi represent a widespread kingdom of pathogenic species for which neither vaccine nor neutralizing antibody therapies are clinically available. Here, using a multi-kingdom antibody profiling (multiKAP) approach, we explore the human antibody repertoires against gut commensal fungi (mycobiota). We identify species preferentially targeted by systemic antibodies in humans, with Candida albicans being the major inducer of antifungal immunoglobulin G (IgG). Fungal colonization of the gut induces germinal center (GC)-dependent B cell expansion in extraintestinal lymphoid tissues and generates systemic antibodies that confer protection against disseminated C. albicans or C. auris infection. Antifungal IgG production depends on the innate immunity regulator CARD9 and CARD9+CX3CR1+ macrophages. In individuals with invasive candidiasis, loss-of-function mutations in CARD9 are associated with impaired antifungal IgG responses. These results reveal an important role of gut commensal fungi in shaping the human antibody repertoire through CARD9-dependent induction of host-protective antifungal IgG.


Assuntos
Anticorpos Antifúngicos/imunologia , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Imunidade , Imunoglobulina G/imunologia , Micobioma/imunologia , Animais , Linfócitos B/imunologia , Candida albicans/imunologia , Candidíase/imunologia , Candidíase/microbiologia , Fezes/microbiologia , Centro Germinativo/imunologia , Humanos , Camundongos Endogâmicos C57BL , Fagócitos/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Ligação Proteica , Transdução de Sinais
7.
Cell Host Microbe ; 27(5): 823-829.e3, 2020 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-32298656

RESUMO

Fecal microbiota transplantation (FMT) targeting gut microbiota has recently been successfully applied to ulcerative colitis. However, only a subset of patients responds to FMT, and there is a pressing need for biomarkers of responsiveness. Fungi (the mycobiota) represent a highly immunologically reactive component of the gut microbiota. We analyzed samples from a large randomized controlled trial of FMT for ulcerative colitis (UC). High Candida abundance pre-FMT was associated with a clinical response, whereas decreased Candida abundance post-FMT was indicative of ameliorated disease severity. High pre-FMT Candida was associated with increased bacterial diversity post-FMT, and the presence of genera was linked to FMT responsiveness. Although we detected elevated anti-Candida antibodies in placebo recipients, this increase was abrogated in FMT recipients. Our data suggest that FMT might reduce Candida to contain pro-inflammatory immunity during intestinal disease and highlight the utility of mycobiota-focused approaches to identify FMT responders prior to therapy initiation.


Assuntos
Colite Ulcerativa/terapia , Transplante de Microbiota Fecal/métodos , Fezes/microbiologia , Fungos , Adulto , Anticorpos Antifúngicos/sangue , Bactérias/genética , Candida , Colite Ulcerativa/microbiologia , Feminino , Fungos/genética , Microbioma Gastrointestinal/fisiologia , Humanos , Masculino , RNA Ribossômico 16S/genética , Resultado do Tratamento
8.
Fungal Genet Biol ; 127: 45-49, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30849443

RESUMO

Gut mycobiota dysbiosis can negatively impact the outcome of several diseases of inflammatory origin, suggesting a role of the mycobiota in influencing the host immunity. However, it is unknown whether the gut mycobiota composition can create an immune environment that would influence the immune response to a newly introduced intestinal fungus. Using ITS1 deep sequencing, we evaluated the mycobiome structure of C57BL/6J mice acquired from Jackson (JAX) or bred in a controlled environment at a dedicated room in our own mouse facility (WCM-CE) for several generations. We found that C57BL/6J mice from these segregated mouse colonies harbor dramatically different mycobiota. To assess whether the mycobiota make up can influence immune responses to colonization with a fungus foreign to the murine GI tract, we colonized JAX and WCM-CE mice with the human commensal C. albicans and measured Th17 responses in the gut. We found that independent of mycobiota composition, mice produced strong Th17 responses to gastrointestinal C. albicans colonization. Our data suggest that different mouse colonies can carry dramatically different mycobiota. Nevertheless, strong Th17 responses to a newly introduced opportunistic commensal fungus are potently induced independent of the mycobiota background in this experimental setting.


Assuntos
Fungos/classificação , Trato Gastrointestinal/microbiologia , Micobioma , Simbiose , Células Th17/imunologia , Animais , DNA Intergênico/genética , DNA Ribossômico/genética , Fungos/fisiologia , Sequenciamento de Nucleotídeos em Larga Escala , Interações Hospedeiro-Patógeno , Camundongos , Camundongos Endogâmicos C57BL
9.
Cell Host Microbe ; 24(6): 847-856.e4, 2018 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-30503509

RESUMO

Sensing of the gut microbiota, including fungi, regulates mucosal immunity. Whether fungal sensing in the gut can influence immunity at other body sites is unknown. Here we show that fluconazole-induced gut fungal dysbiosis has persistent effects on allergic airway disease in a house dust mite challenge model. Mice with a defined community of bacteria, but lacking intestinal fungi were not susceptible to fluconazole-induced dysbiosis, while colonization with a fungal mixture recapitulated the detrimental effects. Gut-resident mononuclear phagocytes (MNPs) expressing the fractalkine receptor CX3CR1 were essential for the effect of gut fungal dysbiosis on peripheral immunity. Depletion of CX3CR1+ MNPs or selective inhibition of Syk signaling downstream of fungal sensing in these cells ameliorated lung allergy. These results indicate that disruption of intestinal fungal communities can have persistent effects on peripheral immunity and aggravate disease severity through fungal sensing by gut-resident CX3CR1+ MNPs.


Assuntos
Disbiose , Hipersensibilidade , Animais , Receptor 1 de Quimiocina CX3C , Fungos , Intestinos , Camundongos , Fagócitos
10.
Sci Transl Med ; 10(461)2018 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-30282696

RESUMO

Gut homing CD4+ T cells expressing the integrin α4ß7 are early viral targets and contribute to HIV-1 pathogenesis, likely by seeding the gastrointestinal (GI) tract with HIV. Although simianized anti-α4ß7 monoclonal antibodies have shown promise in preventing or attenuating the disease course of simian immunodeficiency virus in nonhuman primate studies, the mechanisms of drug action remain elusive. We present a cohort of individuals with mild inflammatory bowel disease and concomitant HIV-1 infection receiving anti-α4ß7 treatment. By sampling the immune inductive and effector sites of the GI tract, we have discovered that anti-α4ß7 therapy led to a significant and unexpected attenuation of lymphoid aggregates, most notably in the terminal ileum. Given that lymphoid aggregates serve as important sanctuary sites for maintaining viral reservoirs, their attrition by anti-α4ß7 therapy has important implications for HIV-1 therapeutics and eradication efforts and defines a rational basis for the use of anti-α4ß7 therapy in HIV-1 infection.


Assuntos
Trato Gastrointestinal/patologia , Trato Gastrointestinal/virologia , Infecções por HIV/terapia , Integrinas/antagonistas & inibidores , Tecido Linfoide/patologia , Adulto , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Linfócitos B/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Feminino , Infecções por HIV/sangue , Infecções por HIV/imunologia , Humanos , Integrinas/metabolismo , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Subpopulações de Linfócitos/efeitos dos fármacos , Subpopulações de Linfócitos/imunologia , Masculino , Pessoa de Meia-Idade , Splicing de RNA/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
11.
Science ; 359(6372): 232-236, 2018 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-29326275

RESUMO

Intestinal fungi are an important component of the microbiota, and recent studies have unveiled their potential in modulating host immune homeostasis and inflammatory disease. Nonetheless, the mechanisms governing immunity to gut fungal communities (mycobiota) remain unknown. We identified CX3CR1+ mononuclear phagocytes (MNPs) as being essential for the initiation of innate and adaptive immune responses to intestinal fungi. CX3CR1+ MNPs express antifungal receptors and activate antifungal responses in a Syk-dependent manner. Genetic ablation of CX3CR1+ MNPs in mice led to changes in gut fungal communities and to severe colitis that was rescued by antifungal treatment. In Crohn's disease patients, a missense mutation in the gene encoding CX3CR1 was identified and found to be associated with impaired antifungal responses. These results unravel a role of CX3CR1+ MNPs in mediating interactions between intestinal mycobiota and host immunity at steady state and during inflammatory disease.


Assuntos
Receptor 1 de Quimiocina CX3C/análise , Receptor 1 de Quimiocina CX3C/genética , Candida albicans/imunologia , Microbioma Gastrointestinal/imunologia , Intestinos/microbiologia , Micobioma/imunologia , Fagócitos/imunologia , Animais , Anticorpos Antifúngicos/biossíntese , Anticorpos Antifúngicos/sangue , Candida albicans/crescimento & desenvolvimento , Colite/tratamento farmacológico , Colite/microbiologia , Doença de Crohn/genética , Doença de Crohn/imunologia , Células Dendríticas/imunologia , Microbioma Gastrointestinal/fisiologia , Humanos , Imunidade nas Mucosas , Imunoglobulina G/biossíntese , Imunoglobulina G/sangue , Intestinos/imunologia , Camundongos , Mutação de Sentido Incorreto , Micobioma/fisiologia , Fagócitos/microbiologia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia
12.
Phys Rev E Stat Nonlin Soft Matter Phys ; 76(5 Pt 1): 051903, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18233683

RESUMO

Much effort has been devoted to developing analysis methods of subdural electroencephalogram and depth electrode recordings of epileptic patients being evaluated for surgical resection. The general approach is to investigate the brain activity at different locations as recorded by the different electrodes in an attempt to localize the epileptogenic focus or foci. Currently, most of the methods are based on the notion that epileptogenic brain activity is associated with changes in synchronization and in complexity. Here we present a method that is based on the temporal dynamics combined with the spectral distribution of energy in terms of frequency-entropy (FE) templates. The FE templates are based upon maximum information partitioning into a set of frequency bands. The FE template is calculated by wavelet packet decomposition followed by an application of the best basis algorithm minimizing the entropy cost function. A comparison between two FE templates is performed by a special quantitative similarity measure according to the overlap in the partitioning into frequency bands and weighted by the bands' entropy. For localization of the epileptogenic foci, the templates of each electrode during the interictal period are compared with a representative template evaluated from the ensemble of all electrodes during the ictal period. We suggest associating the locations that reveal high template similarity to the ictal template with the epileptogenic foci. To test the method and the underlying assumptions, we perform retrospective analysis of the recorded brain activity, from both grid and depth electrodes, from 11 patients suffering from medically intractable epilepsy. Application of the ictal-interictal FE template similarity analysis revealed regions in the epileptic brain in which the interictal characteristics are highly similar to those of the ictal period. To asses the foci we compared the interictal templates of the different electrodes to each other, forming interelectrode similarity matrices. Investigation of these similarity matrices revealed the existence of a single distinct subcluster of electrodes with high interelectrode similarity in the brain activity of seven patients (type-I activity), and the existence of multiple high interelectrode similarity subclusters in the activity of four patients (type-II activity). Comparisons of the analysis results to the medical presurgical evaluations and the outcomes of the surgical resections suggest that the method may be helpful in the chronic evaluation of the epileptogenic zone before operation, and in some cases (type-I activity) without the need to wait for seizures to occur.


Assuntos
Mapeamento Encefálico/métodos , Encéfalo/fisiopatologia , Diagnóstico por Computador/métodos , Eletroencefalografia/métodos , Epilepsia/diagnóstico , Epilepsia/fisiopatologia , Rede Nervosa/fisiopatologia , Simulação por Computador , Entropia , Humanos , Modelos Neurológicos , Sensibilidade e Especificidade , Processamento de Sinais Assistido por Computador
13.
Phys Rev Lett ; 96(25): 258101, 2006 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-16907347

RESUMO

The various human brain tasks are performed at different locations and time scales. Yet, we discovered the existence of time-invariant (above an essential time scale) partitioning of the brain activity into personal state-specific frequency bands. For that, we perform temporal and ensemble averaging of best wavelet packet bases from multielectrode electroencephalogram recordings. These personal frequency bands provide new templates for quantitative analyses of brain function, e.g., normal versus epileptic activity.


Assuntos
Encéfalo/fisiologia , Eletroencefalografia/métodos , Algoritmos , Córtex Cerebral/fisiologia , Eletrodos , Humanos , Modelos Neurológicos , Espaço Subdural/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...