Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 13: 954985, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36052094

RESUMO

Patients suffering from coronavirus disease-2019 (COVID-19) are susceptible to deadly secondary fungal infections such as COVID-19-associated pulmonary aspergillosis and COVID-19-associated mucormycosis. Despite this clinical observation, direct experimental evidence for severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2)-driven alterations of antifungal immunity is scarce. Using an ex-vivo whole blood stimulation assay, we challenged blood from twelve COVID-19 patients with Aspergillus fumigatus and Rhizopus arrhizus antigens and studied the expression of activation, maturation, and exhaustion markers, as well as cytokine secretion. Compared to healthy controls, T-helper cells from COVID-19 patients displayed increased expression levels of the exhaustion marker PD-1 and weakened A. fumigatus- and R. arrhizus-induced activation. While baseline secretion of proinflammatory cytokines was massively elevated, whole blood from COVID-19 patients elicited diminished release of T-cellular (e.g., IFN-γ, IL-2) and innate immune cell-derived (e.g., CXCL9, CXCL10) cytokines in response to A. fumigatus and R. arrhizus antigens. Additionally, samples from COVID-19 patients showed deficient granulocyte activation by mold antigens and reduced fungal killing capacity of neutrophils. These features of weakened anti-mold immune responses were largely decoupled from COVID-19 severity, the time elapsed since diagnosis of COVID-19, and recent corticosteroid uptake, suggesting that impaired anti-mold defense is a common denominator of the underlying SARS-CoV-2 infection. Taken together, these results expand our understanding of the immune predisposition to post-viral mold infections and could inform future studies of immunotherapeutic strategies to prevent and treat fungal superinfections in COVID-19 patients.


Assuntos
COVID-19 , Corticosteroides/uso terapêutico , Aspergillus fumigatus , Citocinas/metabolismo , Humanos , SARS-CoV-2
2.
Front Microbiol ; 13: 896145, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35783442

RESUMO

All Aspergillus species form phialidic conidia (PC) when the mycelium is in contact with the air. These small, asexual spores are ideally suited for an airborne dissemination in the environment. Aspergillus terreus and a few closely related species from section Terrei can additionally generate accessory conidia (AC) that directly emerge from the hyphal surface. In this study, we have identified galactomannan as a major surface antigen on AC that is largely absent from the surface of PC. Galactomannan is homogeneously distributed over the entire surface of AC and even detectable on nascent AC present on the hyphal surface. In contrast, ß-glucans are only accessible in distinct structures that occur after separation of the conidia from the hyphal surface. During germination, AC show a very limited isotropic growth that has no detectable impact on the distribution of galactomannan. The AC of the strain used in this study germinate much faster than the corresponding PC, and they are more sensitive to desiccation than PC. During infection of murine J774 macrophages, AC are readily engulfed and trigger a strong tumor necrosis factor-alpha (TNFα) response. Both processes are not hampered by the presence of laminarin, which indicates that ß-glucans only play a minor role in these interactions. In the phagosome, we observed that galactomannan, but not ß-glucan, is released from the conidial surface and translocates to the host cell cytoplasm. AC persist in phagolysosomes, and many of them initiate germination within 24 h. In conclusion, we have identified galactomannan as a novel and major antigen on AC that clearly distinguishes them from PC. The role of this fungal-specific carbohydrate in the interactions with the immune system remains an open issue that needs to be addressed in future research.

3.
Front Immunol ; 12: 659752, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34122417

RESUMO

Aspergillus fumigatus causes life-threatening opportunistic infections in immunocompromised patients. As therapeutic outcomes of invasive aspergillosis (IA) are often unsatisfactory, the development of targeted immunotherapy remains an important goal. Linking the innate and adaptive immune system, dendritic cells are pivotal in anti-Aspergillus defense and have generated interest as a potential immunotherapeutic approach in IA. While monocyte-derived dendritic cells (moDCs) require ex vivo differentiation, antigen-pulsed primary myeloid dendritic cells (mDCs) may present a more immediate platform for immunotherapy. To that end, we compared the response patterns and cellular interactions of human primary mDCs and moDCs pulsed with an A. fumigatus lysate and two A. fumigatus proteins (CcpA and Shm2) in a serum-free, GMP-compliant medium. CcpA and Shm2 triggered significant upregulation of maturation markers in mDCs and, to a lesser extent, moDCs. Furthermore, both A. fumigatus proteins elicited the release of an array of key pro-inflammatory cytokines including TNF-α, IL-1ß, IL-6, IL-8, and CCL3 from both DC populations. Compared to moDCs, CcpA- and Shm2-pulsed mDCs exhibited greater expression of MHC class II antigens and stimulated stronger proliferation and IFN-γ secretion from autologous CD4+ and CD8+ T-cells. Moreover, supernatants of CcpA- and Shm2-pulsed mDCs significantly enhanced the oxidative burst in allogeneic neutrophils co-cultured with A. fumigatus germ tubes. Taken together, our in vitro data suggest that ex vivo CcpA- and Shm2-pulsed primary mDCs have the potential to be developed into an immunotherapeutic approach to tackle IA.


Assuntos
Aspergillus fumigatus/imunologia , Células Dendríticas/imunologia , Proteínas Fúngicas/imunologia , Ativação Linfocitária/imunologia , Explosão Respiratória/imunologia , Linfócitos T/imunologia , Aspergilose/imunologia , Aspergilose/metabolismo , Aspergilose/microbiologia , Aspergillus fumigatus/metabolismo , Aspergillus fumigatus/fisiologia , Diferenciação Celular/imunologia , Células Cultivadas , Citocinas/imunologia , Citocinas/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/microbiologia , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Monócitos/imunologia , Linfócitos T/metabolismo , Linfócitos T/microbiologia
4.
Sci Rep ; 11(1): 8000, 2021 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-33846501

RESUMO

Here, we present the Oxford Cognitive Screen-Plus, a computerised tablet-based screen designed to briefly assess domain-general cognition and provide more fine-grained measures of memory and executive function. The OCS-Plus was designed to sensitively screen for cognitive impairments and provide a differentiation between memory and executive deficits. The OCS-Plus contains 10 subtasks and requires on average 24 min to complete. In this study, 320 neurologically healthy ageing participants (age M = 62.66, SD = 13.75) from three sites completed the OCS-Plus. The convergent validity of this assessment was established in comparison to the ACE-R, CERAD and Rey-Osterrieth. Divergent validity was established through comparison with the BDI and tests measuring divergent cognitive domains. Internal consistency of each subtask was evaluated, and test-retest reliability was determined. We established the normative impairment cut-offs for each of the subtasks. Predicted convergent and divergent validity was found, high internal consistency for most measures was also found with the exception of restricted range tasks, as well as strong test-retest reliability, which provided evidence of test stability. Further research demonstrating the use and validity of the OCS-Plus in various clinical populations is required. The OCS-Plus is presented as a standardised cognitive assessment tool, normed and validated in a sample of neurologically healthy participants. The OCS-Plus will be available as an Android App and provides an automated report of domain-general cognitive impairments in executive attention and memory.


Assuntos
Disfunção Cognitiva/diagnóstico , Testes Neuropsicológicos , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Escolaridade , Feminino , Alemanha , Humanos , Masculino , Pessoa de Meia-Idade , Reprodutibilidade dos Testes , Reino Unido , Adulto Jovem
5.
Virulence ; 10(1): 677-694, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31274379

RESUMO

The complement system is pivotal in the defense against invasive disease caused by Neisseria meningitidis (Nme, meningococcus), particularly via the membrane attack complex. Complement activation liberates the anaphylatoxins C3a and C5a, which activate three distinct G-protein coupled receptors, C3aR, C5aR1 and C5aR2 (anaphylatoxin receptors, ATRs). We recently discovered that C5aR1 exacerbates the course of the disease, revealing a downside of complement in Nme sepsis. Here, we compared the roles of all three ATRs during mouse nasal colonization, intraperitoneal infection and human whole blood infection with Nme. Deficiency of complement or ATRs did not alter nasal colonization, but significantly affected invasive disease: Compared to WT mice, the disease was aggravated in C3ar-/- mice, whereas C5ar1-/- and C5ar2-/- mice showed increased resistance to meningococcal sepsis. Surprisingly, deletion of either of the ATRs resulted in lower cytokine/chemokine responses, irrespective of the different susceptibilities of the mice. This was similar in ex vivo human whole blood infection using ATR inhibitors. Neutrophil responses to Nme were reduced in C5ar1-/- mouse blood. Upon stimulation with C5a plus Nme, mouse macrophages displayed reduced phosphorylation of ERK1/2, when C5aR1 or C5aR2 were ablated or inhibited, suggesting that both C5a-receptors prime an initial macrophage response to Nme. Finally, in vivo blockade of C5aR1 alone (PMX205) or along with C5aR2 (A8Δ71-73) resulted in ameliorated disease, whereas neither antagonizing C3aR (SB290157) nor its activation with a "super-agonist" peptide (WWGKKYRASKLGLAR) demonstrated a benefit. Thus, C5aR1 and C5aR2 augment disease pathology and are interesting targets for treatment, whereas C3aR is protective in experimental meningococcal sepsis.


Assuntos
Infecções Meningocócicas/imunologia , Neisseria meningitidis/imunologia , Receptor da Anafilatoxina C5a/imunologia , Receptores de Complemento/imunologia , Anafilatoxinas/imunologia , Animais , Quimiocinas/imunologia , Citocinas/imunologia , Humanos , Macrófagos/imunologia , Macrófagos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neisseria meningitidis/patogenicidade , Neutrófilos/imunologia , Neutrófilos/microbiologia , Receptor da Anafilatoxina C5a/genética , Receptores de Complemento/genética , Sepse
6.
Sci Rep ; 9(1): 2736, 2019 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-30804422

RESUMO

Although usually asymptomatically colonizing the human nasopharynx, the Gram-negative bacterium Neisseria meningitidis (meningococcus) can spread to the blood stream and cause invasive disease. For survival in blood, N. meningitidis evades the complement system by expression of a polysaccharide capsule and surface proteins sequestering the complement regulator factor H (fH). Meningococcal strains belonging to the sequence type (ST-) 41/44 clonal complex (cc41/44) cause a major proportion of serogroup B meningococcal disease worldwide, but they are also common in asymptomatic carriers. Proteome analysis comparing cc41/44 isolates from invasive disease versus carriage revealed differential expression levels of the outer membrane protein NspA, which binds fH. Deletion of nspA reduced serum resistance and NspA expression correlated with fH sequestration. Expression levels of NspA depended on the length of a homopolymeric tract in the nspA promoter: A 5-adenosine tract dictated low NspA expression, whereas a 6-adenosine motif guided high NspA expression. Screening German cc41/44 strain collections revealed the 6-adenosine motif in 39% of disease isolates, but only in 3.4% of carriage isolates. Thus, high NspA expression is associated with disease, but not strictly required. The 6-adenosine nspA promoter is most common to the cc41/44, but is also found in other hypervirulent clonal complexes.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Meningite Meningocócica/metabolismo , Neisseria meningitidis Sorogrupo B/fisiologia , Proteínas da Membrana Bacteriana Externa/genética , Fator H do Complemento/metabolismo , Deleção de Genes , Interações Hospedeiro-Patógeno , Humanos , Neisseria meningitidis Sorogrupo B/genética , Poli A/genética , Regiões Promotoras Genéticas , Transcriptoma
7.
Pathog Dis ; 76(8)2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30476070

RESUMO

The complement system is the primary innate immune determinant protecting against invasive diseases caused by the Gram-negative bacterium Neisseria meningitidis (Nme, meningococcus), as evidenced by the extreme susceptibility of individuals with complement deficiencies. In contrast, the role of phagocytes such as neutrophils is much less well understood, although they are recruited in great numbers to the cerebrospinal fluid during meningococcal meningitis. Here, we consider the interaction of Nme with primary human neutrophils using either purified cells or a whole blood model of infection. We found that neutrophils are capable of non-opsonic uptake and killing of different Nme strains. However, in the presence of immune serum featuring active complement, Nme association is strongly increased, whereas this is not the case in heat-inactivated immune serum. Blockade of complement at the level of C3 using the inhibitor compstatin Cp20 reduces the uptake dramatically. In addition, purified neutrophils did not mount an oxidative burst towards Nme unless complement was added and, vice versa, the oxidative burst was strongly reduced in whole blood upon complement inhibition. In contrast, there was no significant impact of complement on neutrophil degranulation or IL-8 secretion. Taken together, neutrophils require complement activation in order to mount a full response towards Nme.


Assuntos
Proteínas do Sistema Complemento/metabolismo , Fatores Imunológicos/metabolismo , Meningite Meningocócica/fisiopatologia , Neisseria meningitidis/imunologia , Neutrófilos/imunologia , Neutrófilos/microbiologia , Adulto , Voluntários Saudáveis , Humanos , Viabilidade Microbiana/efeitos dos fármacos , Modelos Biológicos , Explosão Respiratória , Adulto Jovem
8.
Sci Rep ; 8(1): 10225, 2018 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-29977064

RESUMO

Neisseria meningitidis (meningococcus) causes invasive diseases such as meningitis or septicaemia. Ex vivo infection of human whole blood is a valuable tool to study meningococcal virulence factors and the host innate immune responses. In order to consider effects of cellular mediators, the coagulation cascade must be inhibited to avoid clotting. There is considerable variation in the anticoagulants used among studies of N. meningitidis whole blood infections, featuring citrate, heparin or derivatives of hirudin, a polypeptide from leech saliva. Here, we compare the influence of these three different anticoagulants, and additionally Mg/EGTA, on host innate immune responses as well as on viability of N. meningitidis strains isolated from healthy carriers and disease cases, reflecting different sequence types and capsule phenotypes. We found that the anticoagulants significantly impact on cellular responses and, strain-dependently, also on bacterial survival. Hirudin does not inhibit complement and is therefore superior over the other anticoagulants; indeed hirudin-plasma most closely reflects the characteristics of serum during N. meningitidis infection. We further demonstrate the impact of heparin on complement activation on N. meningitidis and its consequences on meningococcal survival in immune sera, which appears to be independent of the heparin binding antigens Opc and NHBA.


Assuntos
Anticoagulantes/farmacologia , Imunidade Inata/efeitos dos fármacos , Infecções Meningocócicas/imunologia , Neisseria meningitidis/crescimento & desenvolvimento , Animais , Proteínas da Membrana Bacteriana Externa/metabolismo , Células Sanguíneas/imunologia , Células Sanguíneas/microbiologia , Proteínas de Transporte/metabolismo , Ácido Cítrico/farmacologia , Ativação do Complemento/efeitos dos fármacos , Heparina/farmacologia , Hirudinas/farmacologia , Humanos , Viabilidade Microbiana/efeitos dos fármacos , Modelos Biológicos , Neisseria meningitidis/efeitos dos fármacos , Neisseria meningitidis/imunologia
9.
mBio ; 9(1)2018 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-29362231

RESUMO

Sepsis caused by Neisseria meningitidis (meningococcus) is a rapidly progressing, life-threatening disease. Because its initial symptoms are rather unspecific, medical attention is often sought too late, i.e., when the systemic inflammatory response is already unleashed. This in turn limits the success of antibiotic treatment. The complement system is generally accepted as the most important innate immune determinant against invasive meningococcal disease since it protects the host through the bactericidal membrane attack complex. However, complement activation concomitantly liberates the C5a peptide, and it remains unclear whether this potent anaphylatoxin contributes to protection and/or drives the rapidly progressing immunopathogenesis associated with meningococcal disease. Here, we dissected the specific contribution of C5a receptor 1 (C5aR1), the canonical receptor for C5a, using a mouse model of meningococcal sepsis. Mice lacking C3 or C5 displayed susceptibility that was enhanced by >1,000-fold or 100-fold, respectively, consistent with the contribution of these components to protection. In clear contrast, C5ar1-/- mice resisted invasive meningococcal infection and cleared N. meningitidis more rapidly than wild-type (WT) animals. This favorable outcome stemmed from an ameliorated inflammatory cytokine response to N. meningitidis in C5ar1-/- mice in both in vivo and ex vivo whole-blood infections. In addition, inhibition of C5aR1 signaling without interference with the complement bactericidal activity reduced the inflammatory response also in human whole blood. Enticingly, pharmacologic C5aR1 blockade enhanced mouse survival and lowered meningococcal burden even when the treatment was administered after sepsis induction. Together, our findings demonstrate that C5aR1 drives the pathophysiology associated with meningococcal sepsis and provides a promising target for adjunctive therapy.IMPORTANCE The devastating consequences of N. meningitidis sepsis arise due to the rapidly arising and self-propagating inflammatory response that mobilizes antibacterial defenses but also drives the immunopathology associated with meningococcemia. The complement cascade provides innate broad-spectrum protection against infection by directly damaging the envelope of pathogenic microbes through the membrane attack complex and triggers an inflammatory response via the C5a peptide and its receptor C5aR1 aimed at mobilizing cellular effectors of immunity. Here, we consider the potential of separating the bactericidal activities of the complement cascade from its immune activating function to improve outcome of N. meningitidis sepsis. Our findings demonstrate that the specific genetic or pharmacological disruption of C5aR1 rapidly ameliorates disease by suppressing the pathogenic inflammatory response and, surprisingly, allows faster clearance of the bacterial infection. This outcome provides a clear demonstration of the therapeutic benefit of the use of C5aR1-specific inhibitors to improve the outcome of invasive meningococcal disease.


Assuntos
Neisseria meningitidis/imunologia , Neisseria meningitidis/patogenicidade , Receptor da Anafilatoxina C5a/metabolismo , Sepse/microbiologia , Sepse/fisiopatologia , Animais , Atividade Bactericida do Sangue , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Knockout , Receptor da Anafilatoxina C5a/deficiência , Análise de Sobrevida
10.
Infect Immun ; 83(1): 354-63, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25368118

RESUMO

Neisseria meningitidis asymptomatically colonizes the human upper respiratory tract but is also the cause of meningitis and severe septicemia. Carriage or disease evokes an immune response against the infecting strain. Hitherto, we have known little about the breadth of immunity induced by natural carriage of a single strain or its implications for subsequent infectious challenge. In this study, we establish that transgenic mice expressing human CEACAM1 support nasal colonization by a variety of strains of different capsular types. Next, we nasally challenged these mice with either of the N. meningitidis strains H44/76 (serogroup B, ST-32) and 90/18311 (serogroup C, ST-11), while following the induction of strain-specific immunoglobulin. When these antisera were tested for reactivity with a diverse panel of N. meningitidis strains, very low levels of antibody were detected against all meningococcal strains, yet a mutually exclusive "fingerprint" of high-level cross-reactivity toward certain strains became apparent. To test the efficacy of these responses for protection against subsequent challenge, CEACAM1-humanized mice exposed to strain 90/18311 were then rechallenged with different N. meningitidis strains. As expected, the mice were immune to challenge with the same strain and with a closely related ST-11 strain, 38VI, while H44/76 (ST-32) could still colonize these animals. Notably, however, despite the paucity of detectable humoral response against strain 196/87 (ST-32), this strain was unable to colonize the 90/18311-exposed mice. Combined, our data suggest that current approaches may underestimate the actual breadth of mucosal protection gained through natural exposure to N. meningitidis strains.


Assuntos
Anticorpos Antibacterianos/sangue , Antígenos CD/imunologia , Portador Sadio/imunologia , Moléculas de Adesão Celular/imunologia , Infecções Meningocócicas/imunologia , Infecções Meningocócicas/prevenção & controle , Neisseria meningitidis/imunologia , Animais , Antígenos CD/genética , Moléculas de Adesão Celular/genética , Humanos , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA