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.
PLoS One ; 9(3): e90158, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24662889

RESUMO

Clostridium difficile infection (CDI) is frequently diagnosed in recipients of allogeneic hematopoietic stem cell transplantation (allo-HSCT). We characterized early-transplant CDI and its associations, and analyzed serially-collected feces to determine intestinal carriage of toxigenic C. difficile. Fecal specimens were collected longitudinally from 94 patients during allo-HSCT hospitalization, from the start of pre-transplant conditioning until up to 35 days after stem cell infusion. Presence of C. difficile 16S rRNA and tcdB genes was determined. Clinical variables and specimen data were analyzed for association with development of CDI. Historical data from an additional 1144 allo-HSCT patients was also used. Fecal specimens from 37 patients (39%) were found to harbor C. difficile. Early-transplant CDI was diagnosed in 16 of 94 (17%) patients undergoing allo-HSCT; cases were generally mild and resembled non-CDI diarrhea associated with transplant conditioning. CDI was associated with preceding colonization with tcdB-positive C. difficile and conditioning regimen intensity. We found no associations between early-transplant CDI and graft-versus-host disease or CDI later in transplant. CDI occurs with high frequency during the early phase of allo-HSCT, where recipients are pre-colonized with toxigenic C. difficile. During this time, CDI incidence peaks during pre-transplant conditioning, and is correlated to intensity of the treatment. In this unique setting, high rates of CDI may be explained by prior colonization and chemotherapy; however, cases were generally mild and resembled non-infectious diarrhea due to conditioning, raising concerns of misdiagnosis. Further study of this unique population with more discriminating CDI diagnostic tests are warranted.


Assuntos
Clostridioides difficile/fisiologia , Infecções por Clostridium/etiologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Adulto , Idoso , Estudos de Coortes , Determinação de Ponto Final , Fezes/microbiologia , Feminino , Humanos , Intestinos/microbiologia , Masculino , Pessoa de Meia-Idade , Transplante Homólogo/efeitos adversos , Adulto Jovem
2.
Nat Med ; 19(3): 372-7, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23435170

RESUMO

Delayed T cell recovery and restricted T cell receptor (TCR) diversity after allogeneic hematopoietic stem cell transplantation (allo-HSCT) are associated with increased risks of infection and cancer relapse. Technical challenges have limited faithful measurement of TCR diversity after allo-HSCT. Here we combined 5' rapid amplification of complementary DNA ends PCR with deep sequencing to quantify TCR diversity in 28 recipients of allo-HSCT using a single oligonucleotide pair. Analysis of duplicate blood samples confirmed that we accurately determined the frequency of individual TCRs. After 6 months, cord blood-graft recipients approximated the TCR diversity of healthy individuals, whereas recipients of T cell-depleted peripheral-blood stem cell grafts had 28-fold and 14-fold lower CD4(+) and CD8(+) T cell diversities, respectively. After 12 months, these deficiencies had improved for the CD4(+) but not the CD8(+) T cell compartment. Overall, this method provides unprecedented views of T cell repertoire recovery after allo-HSCT and may identify patients at high risk of infection or relapse.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Transplante de Células-Tronco Hematopoéticas , Receptores de Antígenos de Linfócitos T/genética , Sequência de Bases , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Variação Genética , Neoplasias Hematológicas/sangue , Neoplasias Hematológicas/imunologia , Humanos , Recidiva , Análise de Sequência de DNA
3.
Infect Immun ; 81(3): 965-73, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23319552

RESUMO

Bacteria causing infections in hospitalized patients are increasingly antibiotic resistant. Classical infection control practices are only partially effective at preventing spread of antibiotic-resistant bacteria within hospitals. Because the density of intestinal colonization by the highly antibiotic-resistant bacterium vancomycin-resistant Enterococcus (VRE) can exceed 10(9) organisms per gram of feces, even optimally implemented hygiene protocols often fail. Decreasing the density of intestinal colonization, therefore, represents an important approach to limit VRE transmission. We demonstrate that reintroduction of a diverse intestinal microbiota to densely VRE-colonized mice eliminates VRE from the intestinal tract. While oxygen-tolerant members of the microbiota are ineffective at eliminating VRE, administration of obligate anaerobic commensal bacteria to mice results in a billionfold reduction in the density of intestinal VRE colonization. 16S rRNA gene sequence analysis of intestinal bacterial populations isolated from mice that cleared VRE following microbiota reconstitution revealed that recolonization with a microbiota that contains Barnesiella correlates with VRE elimination. Characterization of the fecal microbiota of patients undergoing allogeneic hematopoietic stem cell transplantation demonstrated that intestinal colonization with Barnesiella confers resistance to intestinal domination and bloodstream infection with VRE. Our studies indicate that obligate anaerobic bacteria belonging to the Barnesiella genus enable clearance of intestinal VRE colonization and may provide novel approaches to prevent the spread of highly antibiotic-resistant bacteria.


Assuntos
Bacteroidaceae/fisiologia , Enterococcus faecium/efeitos dos fármacos , Infecções por Bactérias Gram-Positivas/terapia , Intestinos/microbiologia , Resistência a Vancomicina , Animais , DNA Bacteriano , Feminino , Camundongos , Camundongos Endogâmicos C57BL , RNA Bacteriano/genética , RNA Ribossômico 16S/genética
4.
J Exp Med ; 209(8): 1445-56, 2012 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-22826298

RESUMO

The intestinal microbiota contributes to the development of the immune system, and conversely, the immune system influences the composition of the microbiota. Toll-like receptors (TLRs) in the gut recognize bacterial ligands. Although TLR signaling represents a major arm of the innate immune system, the extent to which TLRs influence the composition of the intestinal microbiota remains unclear. We performed deep 16S ribosomal RNA sequencing to characterize the complex bacterial populations inhabiting the ileum and cecum of TLR- and MyD88-deficient mice. The microbiota of MyD88- and TLR-deficient mouse colonies differed markedly, with each colony harboring distinct and distinguishable bacterial populations in the small and large intestine. Comparison of MyD88-, TLR2-, TLR4-, TLR5-, and TLR9-deficient mice and their respective wild-type (WT) littermates demonstrated that the impact of TLR deficiency on the composition of the intestinal microbiota is minimal under homeostatic conditions and after recovery from antibiotic treatment. Thus, differences between TLR-deficient mouse colonies reflected long-term divergence of the microbiota after extended husbandry in isolation from each other. Long-term breeding of isolated mouse colonies results in changes of the intestinal microbiota that are communicated to offspring by maternal transmission, which account for marked compositional differences between WT and mutant mouse strains.


Assuntos
Ceco/imunologia , Íleo/imunologia , Imunidade Inata/imunologia , Metagenoma/imunologia , Receptores Toll-Like/deficiência , Receptores Toll-Like/imunologia , Animais , Antibacterianos/imunologia , Ceco/microbiologia , Feminino , Íleo/microbiologia , Imunidade Inata/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , RNA Ribossômico 16S/genética , RNA Ribossômico 16S/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptores Toll-Like/genética
5.
Clin Infect Dis ; 55(7): 905-14, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22718773

RESUMO

BACKGROUND: Bacteremia is a frequent complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). It is unclear whether changes in the intestinal microbiota during allo-HSCT contribute to the development of bacteremia. We examined the microbiota of patients undergoing allo-HSCT, and correlated microbial shifts with the risk of bacteremia. METHODS: Fecal specimens were collected longitudinally from 94 patients undergoing allo-HSCT, from before transplant until 35 days after transplant. The intestinal microbiota was characterized by 454 pyrosequencing of the V1-V3 region of bacterial 16S ribosomal RNA genes. Microbial diversity was estimated by grouping sequences into operational taxonomic units and calculating the Shannon diversity index. Phylogenetic classification was obtained using the Ribosomal Database Project classifier. Associations of the microbiota with clinical predictors and outcomes were evaluated. RESULTS: During allo-HSCT, patients developed reduced diversity, with marked shifts in bacterial populations inhabiting the gut. Intestinal domination, defined as occupation of at least 30% of the microbiota by a single predominating bacterial taxon, occurred frequently. Commonly encountered dominating organisms included Enterococcus, Streptococcus, and various Proteobacteria. Enterococcal domination was increased 3-fold by metronidazole administration, whereas domination by Proteobacteria was reduced 10-fold by fluoroquinolone administration. As a predictor of outcomes, enterococcal domination increased the risk of Vancomycin-resistant Enterococcus bacteremia 9-fold, and proteobacterial domination increased the risk of gram-negative rod bacteremia 5-fold. CONCLUSIONS: During allo-HSCT, the diversity and stability of the intestinal flora are disrupted, resulting in domination by bacteria associated with subsequent bacteremia. Assessment of fecal microbiota identifies patients at highest risk for bloodstream infection during allo-HCST.


Assuntos
Bacteriemia/epidemiologia , Bacteriemia/microbiologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Adulto , Idoso , Biodiversidade , DNA Bacteriano/química , DNA Bacteriano/genética , DNA Ribossômico/química , DNA Ribossômico/genética , Fezes/microbiologia , Feminino , Humanos , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Filogenia , RNA Ribossômico 16S/genética , Fatores de Risco , Análise de Sequência de DNA
6.
J Exp Med ; 209(5): 903-11, 2012 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-22547653

RESUMO

Despite a growing understanding of the link between intestinal inflammation and resident gut microbes, longitudinal studies of human flora before initial onset of intestinal inflammation have not been reported. Here, we demonstrate in murine and human recipients of allogeneic bone marrow transplantation (BMT) that intestinal inflammation secondary to graft-versus-host disease (GVHD) is associated with major shifts in the composition of the intestinal microbiota. The microbiota, in turn, can modulate the severity of intestinal inflammation. In mouse models of GVHD, we observed loss of overall diversity and expansion of Lactobacillales and loss of Clostridiales. Eliminating Lactobacillales from the flora of mice before BMT aggravated GVHD, whereas reintroducing the predominant species of Lactobacillus mediated significant protection against GVHD. We then characterized gut flora of patients during onset of intestinal inflammation caused by GVHD and found patterns mirroring those in mice. We also identified increased microbial chaos early after allogeneic BMT as a potential risk factor for subsequent GVHD. Together, these data demonstrate regulation of flora by intestinal inflammation and suggest that flora manipulation may reduce intestinal inflammation and improve outcomes for allogeneic BMT recipients.


Assuntos
Biodiversidade , Transplante de Medula Óssea/efeitos adversos , Enterocolite/microbiologia , Doença Enxerto-Hospedeiro/complicações , Metagenoma/genética , Ampicilina , Animais , Sequência de Bases , Sulfato de Dextrana , Enterocolite/etiologia , Enterocolite/patologia , Fezes/microbiologia , Doença Enxerto-Hospedeiro/microbiologia , Bactérias Gram-Positivas/isolamento & purificação , Humanos , Camundongos , Dados de Sequência Molecular , Filogenia , RNA Ribossômico 16S/genética , Análise de Sequência de DNA , Especificidade da Espécie , Transplante Homólogo
7.
Arthritis Rheum ; 64(10): 3083-94, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22576262

RESUMO

OBJECTIVE: To profile the abundance and diversity of subgingival oral microbiota in patients with never-treated, new-onset rheumatoid arthritis (RA). METHODS: Periodontal disease (PD) status, clinical activity, and sociodemographic factors were determined in patients with new-onset RA, patients with chronic RA, and healthy subjects. Multiplexed-454 pyrosequencing was used to compare the composition of subgingival microbiota and establish correlations between the presence/abundance of bacteria and disease phenotypes. Anti-Porphyromonas gingivalis antibody testing was performed to assess prior exposure to the bacterial pathogen P gingivalis. RESULTS: The more advanced forms of periodontitis were already present at disease onset in patients with new-onset RA. The subgingival microbiota observed in patients with new-onset RA was distinct from that found in healthy controls. In most cases, however, these microbial differences could be attributed to the severity of PD and were not inherent to RA. The presence and abundance of P gingivalis were also directly associated with the severity of PD and were not unique to RA. The presence of P gingivalis was not correlated with anti-citrullinated protein antibody (ACPA) titers. Overall exposure to P gingivalis was similar between patients with new-onset RA and controls, observed in 78% of patients and 83% of controls. The presence and abundance of Anaeroglobus geminatus correlated with the presence of ACPAs/rheumatoid factor. Prevotella and Leptotrichia species were the only characteristic taxa observed in patients with new-onset RA irrespective of PD status. CONCLUSION: Patients with new-onset RA exhibited a high prevalence of PD at disease onset, despite their young age and paucity of smoking history. The subgingival microbiota profile in patients with new-onset RA was similar to that in patients with chronic RA and healthy subjects whose PD was of comparable severity. Although colonization with P gingivalis correlated with the severity of PD, overall exposure to P gingivalis was similar among the groups. The role of A geminatus and Prevotella/Leptotrichia species in this process merits further study.


Assuntos
Artrite Reumatoide/microbiologia , Metagenoma , Boca/microbiologia , Periodontite/microbiologia , Adulto , Artrite Reumatoide/complicações , Artrite Reumatoide/imunologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Boca/imunologia , Periodontite/complicações , Periodontite/imunologia , Porphyromonas gingivalis/imunologia , Índice de Gravidade de Doença , Fatores Socioeconômicos , Inquéritos e Questionários
8.
Infect Immun ; 80(1): 62-73, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22006564

RESUMO

Antibiotic-induced changes in the intestinal microbiota predispose mammalian hosts to infection with antibiotic-resistant pathogens. Clostridium difficile is a Gram-positive intestinal pathogen that causes colitis and diarrhea in patients following antibiotic treatment. Clindamycin predisposes patients to C. difficile colitis. Here, we have used Roche-454 16S rRNA gene pyrosequencing to longitudinally characterize the intestinal microbiota of mice following clindamycin treatment in the presence or absence of C. difficile infection. We show that a single dose of clindamycin markedly reduces the diversity of the intestinal microbiota for at least 28 days, with an enduring loss of ca. 90% of normal microbial taxa from the cecum. Loss of microbial complexity results in dramatic sequential expansion and contraction of a subset of bacterial taxa that are minor contributors to the microbial consortium prior to antibiotic treatment. Inoculation of clindamycin-treated mice with C. difficile (VPI 10463) spores results in rapid development of diarrhea and colitis, with a 4- to 5-day period of profound weight loss and an associated 40 to 50% mortality rate. Recovering mice resolve diarrhea and regain weight but remain highly infected with toxin-producing vegetative C. difficile bacteria and, in comparison to the acute stage of infection, have persistent, albeit ameliorated cecal and colonic inflammation. The microbiota of "recovered" mice remains highly restricted, and mice remain susceptible to C. difficile infection at least 10 days following clindamycin, suggesting that resolution of diarrhea and weight gain may result from the activation of mucosal immune defenses.


Assuntos
Antibacterianos/administração & dosagem , Bactérias/efeitos dos fármacos , Clindamicina/administração & dosagem , Infecções por Clostridium/imunologia , Colite/imunologia , Suscetibilidade a Doenças , Trato Gastrointestinal/metabolismo , Animais , Bactérias/classificação , Bactérias/genética , Biodiversidade , Clostridioides difficile/patogenicidade , Infecções por Clostridium/microbiologia , Infecções por Clostridium/mortalidade , Colite/microbiologia , Colite/mortalidade , Diarreia/imunologia , Diarreia/microbiologia , Diarreia/mortalidade , Feminino , Estudos Longitudinais , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência de DNA/métodos , Análise de Sobrevida , Fatores de Tempo
9.
Immunity ; 34(4): 590-601, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21458307

RESUMO

Inflammatory (Ly6C(hi) CCR2+) monocytes provide defense against infections but also contribute to autoimmune diseases and atherosclerosis. Monocytes originate from bone marrow and their entry into the bloodstream requires stimulation of CCR2 chemokine receptor by monocyte chemotactic protein-1 (MCP1). How monocyte emigration from bone marrow is triggered by remote infections remains unclear. We demonstrated that low concentrations of Toll-like receptor (TLR) ligands in the bloodstream drive CCR2-dependent emigration of monocytes from bone marrow. Bone marrow mesenchymal stem cells (MSCs) and their progeny, including CXC chemokine ligand (CXCL)12-abundant reticular (CAR) cells, rapidly expressed MCP1 in response to circulating TLR ligands or bacterial infection and induced monocyte trafficking into the bloodstream. Targeted deletion of MCP1 from MSCs impaired monocyte emigration from bone marrow. Our findings suggest that bone marrow MSCs and CAR cells respond to circulating microbial molecules and regulate bloodstream monocyte frequencies by secreting MCP1 in proximity to bone marrow vascular sinuses.


Assuntos
Medula Óssea/imunologia , Movimento Celular , Células-Tronco Mesenquimais/imunologia , Monócitos/citologia , Monócitos/imunologia , Receptores Toll-Like/imunologia , Animais , Ligantes , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores CCR2/imunologia
10.
Infect Immun ; 79(4): 1498-503, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21245274

RESUMO

Clostridium difficile is a spore-forming bacterium that infects the lower intestinal tract of humans and is the most common known cause of diarrhea among hospitalized patients. Clostridium difficile colitis is mediated by toxins and develops during or following antibiotic administration. We have used a murine model of C. difficile infection, which reproduces the major features of the human disease, to study the effect of innate immune activation on resistance to C. difficile infection. We found that administration of purified Salmonella-derived flagellin, a Toll-like receptor 5 (TLR5) agonist, protects mice from C. difficile colitis by delaying C. difficile growth and toxin production in the colon and cecum. TLR5 stimulation significantly improves pathological changes in the cecum and colon of C. difficile-infected mice and reduces epithelial cell loss. Flagellin treatment reduces epithelial apoptosis in the large intestine, thereby protecting the integrity of the intestinal epithelial barrier during C. difficile infection. We demonstrate that restoring intestinal innate immune tone by TLR stimulation in antibiotic-treated mice ameliorates intestinal inflammation and prevents death from C. difficile colitis, potentially providing an approach to prevent C. difficile-induced pathology.


Assuntos
Clostridioides difficile/imunologia , Enterocolite Pseudomembranosa/imunologia , Receptor 5 Toll-Like/agonistas , Receptor 5 Toll-Like/imunologia , Doença Aguda , Animais , Modelos Animais de Doenças , Enterocolite Pseudomembranosa/patologia , Feminino , Flagelina/imunologia , Flagelina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
11.
Cell Host Microbe ; 6(5): 470-81, 2009 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-19917501

RESUMO

Aspergillus fumigatus, a ubiquitous fungus, causes invasive disease in immunocompromised humans. Although monocytes and antigen-specific CD4 T cells contribute to defense against inhaled fungal spores, how these cells interact during infection remains undefined. Investigating the role of inflammatory monocytes and monocyte-derived dendritic cells during fungal infection, we find that A. fumigatus infection induces an influx of chemokine receptor CCR2- and Ly6C-expressing inflammatory monocytes into lungs and draining lymph nodes. Depletion of CCR2(+) cells reduced A. fumigatus conidial transport from lungs to draining lymph nodes, abolished CD4 T cell priming following respiratory challenge, and impaired pulmonary fungal clearance. In contrast, depletion of CCR2(+)Ly6C(hi) monocytes during systemic fungal infection did not prevent CD4 T cell priming in the spleen. Our findings demonstrate that pulmonary CD4 T cell responses to inhaled spores require CCR2(+)Ly6C(hi) monocytes and their derivatives, revealing a compartmentally restricted function for these cells in adaptive respiratory immune responses.


Assuntos
Apresentação de Antígeno , Antígenos Ly/imunologia , Aspergillus fumigatus , Linfócitos T CD4-Positivos/imunologia , Movimento Celular/imunologia , Células Dendríticas/imunologia , Monócitos/imunologia , Aspergilose Pulmonar/imunologia , Receptores CCR2/imunologia , Animais , Antígenos Ly/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Células Dendríticas/metabolismo , Inflamação , Pulmão/imunologia , Linfonodos/imunologia , Camundongos , Monócitos/metabolismo , Receptores CCR2/metabolismo , Baço/imunologia
12.
J Infect Dis ; 200(4): 647-56, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19591573

RESUMO

Alveolar macrophages and neutrophils mediate innate immune defense against the opportunistic fungal pathogen Aspergillus fumigatus and are believed to be essential for host survival following inhalation of fungal spores (conidia). Although alveolar macrophages are postulated to kill inhaled conidia and neutrophils are believed to act against hyphae, the relative contribution of alveolar macrophages and neutrophils to early defense against A. fumigatus remain incompletely defined. To more precisely characterize the contributions of alveolar macrophages and neutrophils in antifungal host defense, we selectively depleted each cell population at different times following pulmonary challenge with conidia. Mice depleted of alveolar macrophages prior to pulmonary A. fumigatus infection recruited neutrophils normally and restricted hyphal tissue invasion. In contrast, neutrophil depletion prior to or within 3 h after infection was associated with high mortality. Neutrophil depletion at later time points, however, was associated with nearly normal survival rates. Our studies suggest that neutrophils, but not alveolar macrophages, provide essential anticonidial defense and that a brief period of influx into the respiratory tree is sufficient to prevent conidial germination and invasive disease.


Assuntos
Aspergillus fumigatus , Macrófagos Alveolares/fisiologia , Neutrófilos/fisiologia , Aspergilose Pulmonar/imunologia , Animais , Pulmão/citologia , Camundongos , Camundongos Endogâmicos C57BL , Aspergilose Pulmonar/microbiologia , Fatores de Tempo
13.
J Immunol ; 183(1): 631-41, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19542472

RESUMO

Aspergillus fumigatus, a common environmental fungus, can cause lethal invasive infections in immunocompromised hosts. In immunocompetent individuals, however, inhaled A. fumigatus spores prime CD4(+) T cells and activate immune responses that prevent invasive infection. Calibration of inflammatory responses to levels that prevent fungal invasion without inducing collateral tissue damage is essential for host survival, but the underlying regulatory mechanisms remain undefined. Although IL-10 is a validated regulatory cytokine that suppresses immune responses, and IL-10 deficiency or blockade generally enhances immune responses, we find that A. fumigatus-specific T cell frequencies are markedly reduced in airways of IL-10-deficient mice. T cell priming, proliferation, and survival were unaffected by IL-10 deficiency and did not account for decreased frequencies of A. fumigatus-specific T cells in the airways of IL-10-deficient mice. Instead, IL-10 deficiency results in redistribution of A. fumigatus-specific T cells from infected lungs to the gut, a process that is reversed by antibiotic-mediated depletion of intestinal microbes. Our studies demonstrate that disregulated immune responses in the gut can result in dramatic redistribution of pathogen-specific T cells within the host.


Assuntos
Aspergilose/imunologia , Aspergilose/patologia , Aspergillus fumigatus/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/microbiologia , Epitopos de Linfócito T/imunologia , Interleucina-10/deficiência , Animais , Aspergilose/genética , Linfócitos T CD4-Positivos/patologia , Movimento Celular/imunologia , Células Cultivadas , Interleucina-10/genética , Intestinos/imunologia , Intestinos/microbiologia , Intestinos/patologia , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Linfonodos/imunologia , Linfonodos/microbiologia , Linfonodos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...