Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 228
Filtrar
1.
Nat Commun ; 15(1): 4326, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38773113

RESUMO

Resolving inflammation is thought to return the affected tissue back to homoeostasis but recent evidence supports a non-linear model of resolution involving a phase of prolonged immune activity. Here we show that within days following resolution of Streptococcus pneumoniae-triggered lung inflammation, there is an influx of antigen specific lymphocytes with a memory and tissue-resident phenotype as well as macrophages bearing alveolar or interstitial phenotype. The transcriptome of these macrophages shows enrichment of genes associated with prostaglandin biosynthesis and genes that drive T cell chemotaxis and differentiation. Therapeutic depletion of post-resolution macrophages, inhibition of prostaglandin E2 (PGE2) synthesis or treatment with an EP4 antagonist, MF498, reduce numbers of lung CD4+/CD44+/CD62L+ and CD4+/CD44+/CD62L-/CD27+ T cells as well as their expression of the α-integrin, CD103. The T cells fail to reappear and reactivate upon secondary challenge for up to six weeks following primary infection. Concomitantly, EP4 antagonism through MF498 causes accumulation of lung macrophages and marked tissue fibrosis. Our study thus shows that PGE2 signalling, predominantly via EP4, plays an important role during the second wave of immune activity following resolution of inflammation. This secondary immune activation drives local tissue-resident T cell development while limiting tissue injury.


Assuntos
Dinoprostona , Modelos Animais de Doenças , Pulmão , Macrófagos , Camundongos Endogâmicos C57BL , Pneumonia Pneumocócica , Receptores de Prostaglandina E Subtipo EP4 , Streptococcus pneumoniae , Animais , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/patologia , Pneumonia Pneumocócica/microbiologia , Pneumonia Pneumocócica/metabolismo , Camundongos , Dinoprostona/metabolismo , Streptococcus pneumoniae/imunologia , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Receptores de Prostaglandina E Subtipo EP4/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Pulmão/imunologia , Pulmão/patologia , Pulmão/microbiologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Cadeias alfa de Integrinas/metabolismo , Cadeias alfa de Integrinas/genética , Feminino , Antígenos CD/metabolismo , Antígenos CD/genética , Linfócitos T/imunologia
2.
JCI Insight ; 9(8)2024 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-38646937

RESUMO

Sepsis is a leading cause of mortality worldwide, and pneumonia is the most common cause of sepsis in humans. Low levels of high-density lipoprotein cholesterol (HDL-C) levels are associated with an increased risk of death from sepsis, and increasing levels of HDL-C by inhibition of cholesteryl ester transfer protein (CETP) decreases mortality from intraabdominal polymicrobial sepsis in APOE*3-Leiden.CETP mice. Here, we show that treatment with the CETP inhibitor (CETPi) anacetrapib reduced mortality from Streptococcus pneumoniae-induced sepsis in APOE*3-Leiden.CETP and APOA1.CETP mice. Mechanistically, CETP inhibition reduced the host proinflammatory response via attenuation of proinflammatory cytokine transcription and release. This effect was dependent on the presence of HDL, leading to attenuation of immune-mediated organ damage. In addition, CETP inhibition promoted monocyte activation in the blood prior to the onset of sepsis, resulting in accelerated macrophage recruitment to the lung and liver. In vitro experiments demonstrated that CETP inhibition significantly promoted the activation of proinflammatory signaling in peripheral blood mononuclear cells and THP1 cells in the absence of HDL; this may represent a mechanism responsible for improved bacterial clearance during sepsis. These findings provide evidence that CETP inhibition represents a potential approach to reduce mortality from pneumosepsis.


Assuntos
Proteínas de Transferência de Ésteres de Colesterol , Monócitos , Streptococcus pneumoniae , Animais , Feminino , Humanos , Camundongos , Apolipoproteína E3/metabolismo , Proteínas de Transferência de Ésteres de Colesterol/antagonistas & inibidores , Proteínas de Transferência de Ésteres de Colesterol/metabolismo , HDL-Colesterol/sangue , HDL-Colesterol/metabolismo , Modelos Animais de Doenças , Macrófagos/imunologia , Macrófagos/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/mortalidade , Pneumonia Pneumocócica/metabolismo , Pneumonia Pneumocócica/microbiologia , Sepse/imunologia , Sepse/mortalidade , Sepse/microbiologia , Sepse/metabolismo , Streptococcus pneumoniae/imunologia , Células THP-1
3.
Anesthesiology ; 136(2): 293-313, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34965287

RESUMO

BACKGROUND: Mechanical ventilation for pneumonia may contribute to lung injury due to factors that include mitochondrial dysfunction, and mesenchymal stem cells may attenuate injury. This study hypothesized that mechanical ventilation induces immune and mitochondrial dysfunction, with or without pneumococcal pneumonia, that could be mitigated by mesenchymal stem cells alone or combined with antibiotics. METHODS: Male rabbits underwent protective mechanical ventilation (8 ml/kg tidal volume, 5 cm H2O end-expiratory pressure) or adverse mechanical ventilation (20 ml/kg tidal-volume, zero end-expiratory pressure) or were allowed to breathe spontaneously. The same settings were then repeated during pneumococcal pneumonia. Finally, infected animals during adverse mechanical ventilation received human umbilical cord-derived mesenchymal stem cells (3 × 106/kg, intravenous) and/or ceftaroline (20 mg/kg, intramuscular) or sodium chloride, 4 h after pneumococcal challenge. Twenty-four-hour survival (primary outcome), lung injury, bacterial burden, immune and mitochondrial dysfunction, and lung transcriptomes (secondary outcomes) were assessed. RESULTS: High-pressure adverse mechanical ventilation reduced the survival of infected animals (0%; 0 of 7) compared with spontaneous breathing (100%; 7 of 7) and protective mechanical ventilation (86%; 6 of 7; both P < 0.001), with higher lung pathology scores (median [interquartile ranges], 5.5 [4.5 to 7.0] vs. 12.6 [12.0 to 14.0]; P = 0.046), interleukin-8 lung concentrations (106 [54 to 316] vs. 804 [753 to 868] pg/g of lung; P = 0.012), and alveolar mitochondrial DNA release (0.33 [0.28 to 0.36] vs. 0.98 [0.76 to 1.21] ng/µl; P < 0.001) compared with infected spontaneously breathing animals. Survival (0%; 0 of 7; control group) was improved by mesenchymal stem cells (57%; 4 of 7; P = 0.001) or ceftaroline alone (57%; 4 of 7; P < 0.001) and improved even more with a combination treatment (86%; 6 of 7; P < 0.001). Mesenchymal stem cells reduced lung pathology score (8.5 [7.0 to 10.5] vs. 12.6 [12.0 to 14.0]; P = 0.043) and alveolar mitochondrial DNA release (0.39 (0.34 to 0.65) vs. 0.98 (0.76 to 1.21) ng/µl; P = 0.025). Mesenchymal stem cells combined with ceftaroline reduced interleukin-8 lung concentrations (665 [595 to 795] vs. 804 [753 to 868] pg/g of lung; P = 0.007) compared to ceftaroline alone. CONCLUSIONS: In this preclinical study, mesenchymal stem cells improved the outcome of rabbits with pneumonia and high-pressure mechanical ventilation by correcting immune and mitochondrial dysfunction and when combined with the antibiotic ceftaroline was synergistic in mitigating lung inflammation.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Imunidade Celular/fisiologia , Mitocôndrias/imunologia , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/terapia , Respiração Artificial/efeitos adversos , Animais , Masculino , Células-Tronco Mesenquimais/fisiologia , Mitocôndrias/metabolismo , Pneumonia Pneumocócica/metabolismo , Estudos Prospectivos , Coelhos , Distribuição Aleatória
4.
Sci Rep ; 11(1): 20609, 2021 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-34663857

RESUMO

Seasonal Influenza A virus (IAV) infections can promote dissemination of upper respiratory tract commensals such as Streptococcus pneumoniae to the lower respiratory tract resulting in severe life-threatening pneumonia. Here, we aimed to compare innate immune responses in the lungs of healthy colonized and non-colonized mice after IAV challenge at the initial asymptomatic stage of infection. Responses during a severe bacterial pneumonia were profiled for comparison. Cytokine and innate immune cell imprints of the lungs were analyzed. Irrespective of the colonization status, mild H1N1 IAV infection was characterized by a bi-phasic disease progression resulting in full recovery of the animals. Already at the asymptomatic stage of viral infection, the pro-inflammatory cytokine response was as high as in pneumococcal pneumonia. Flow cytometry analyses revealed an early influx of inflammatory monocytes into the lungs. Neutrophil influx was mostly limited to bacterial infections. The majority of cells, except monocytes, displayed an activated phenotype characterized by elevated CCR2 and MHCII expression. In conclusion, we show that IAV challenge of colonized healthy mice does not automatically result in severe co-infection. However, a general local inflammatory response was noted at the asymptomatic stage of infection irrespective of the infection type.


Assuntos
Imunidade Inata/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções Pneumocócicas/imunologia , Animais , Portador Sadio/imunologia , Coinfecção/virologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H1N1/patogenicidade , Vírus da Influenza A/imunologia , Vírus da Influenza A/patogenicidade , Pulmão/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/metabolismo , Infecções por Orthomyxoviridae/virologia , Infecções Pneumocócicas/complicações , Pneumonia Bacteriana , Pneumonia Pneumocócica/imunologia , Cultura Primária de Células , Infecções Respiratórias/imunologia , Infecções Respiratórias/virologia , Streptococcus pneumoniae/patogenicidade
5.
J Immunol ; 207(5): 1357-1370, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34380651

RESUMO

Zinc (Zn) is required for proper immune function and host defense. Zn homeostasis is tightly regulated by Zn transporters that coordinate biological processes through Zn mobilization. Zn deficiency is associated with increased susceptibility to bacterial infections, including Streptococcus pneumoniae, the most commonly identified cause of community-acquired pneumonia. Myeloid cells, including macrophages and dendritic cells (DCs), are at the front line of host defense against invading bacterial pathogens in the lung and play a critical role early on in shaping the immune response. Expression of the Zn transporter ZIP8 is rapidly induced following bacterial infection and regulates myeloid cell function in a Zn-dependent manner. To what extent ZIP8 is instrumental in myeloid cell function requires further study. Using a novel, myeloid-specific, Zip8 knockout model, we identified vital roles of ZIP8 in macrophage and DC function upon pneumococcal infection. Administration of S. pneumoniae into the lung resulted in increased inflammation, morbidity, and mortality in Zip8 knockout mice compared with wild-type counterparts. This was associated with increased numbers of myeloid cells, cytokine production, and cell death. In vitro analysis of macrophage and DC function revealed deficits in phagocytosis and increased cytokine production upon bacterial stimulation that was, in part, due to increased NF-κB signaling. Strikingly, alteration of myeloid cell function resulted in an imbalance of Th17/Th2 responses, which is potentially detrimental to host defense. These results (for the first time, to our knowledge) reveal a vital ZIP8- and Zn-mediated axis that alters the lung myeloid cell landscape and the host response against pneumococcus.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Células Dendríticas/imunologia , Macrófagos/imunologia , Células Mieloides/imunologia , Pneumonia Pneumocócica/imunologia , Streptococcus pneumoniae/fisiologia , Células Th17/imunologia , Células Th2/imunologia , Animais , Proteínas de Transporte de Cátions/genética , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Fagocitose/genética , Transdução de Sinais
6.
J Clin Invest ; 131(11)2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34060477

RESUMO

Lung-resident memory B cells (BRM cells) are elicited after influenza infections of mice, but connections to other pathogens and hosts - as well as their functional significance - have yet to be determined. We postulate that BRM cells are core components of lung immunity. To test this, we examined whether lung BRM cells are elicited by the respiratory pathogen pneumococcus, are present in humans, and are important in pneumonia defense. Lungs of mice that had recovered from pneumococcal infections did not contain organized tertiary lymphoid organs, but did have plasma cells and noncirculating memory B cells. The latter expressed distinctive surface markers (including CD69, PD-L2, CD80, and CD73) and were poised to secrete antibodies upon stimulation. Human lungs also contained B cells with a resident memory phenotype. In mice recovered from pneumococcal pneumonia, depletion of PD-L2+ B cells, including lung BRM cells, diminished bacterial clearance and the level of pneumococcus-reactive antibodies in the lung. These data define lung BRM cells as a common feature of pathogen-experienced lungs and provide direct evidence of a role for these cells in pulmonary antibacterial immunity.


Assuntos
Linfócitos B/imunologia , Memória Imunológica , Pulmão/imunologia , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/prevenção & controle , Streptococcus pneumoniae/imunologia , Animais , Antígenos de Diferenciação/imunologia , Linfócitos B/patologia , Humanos , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Transgênicos , Pneumonia Pneumocócica/microbiologia , Pneumonia Pneumocócica/patologia
7.
Biochem J ; 478(8): 1485-1509, 2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33881487

RESUMO

Carbohydrate-binding antibodies play diverse and critical roles in human health. Endogenous carbohydrate-binding antibodies that recognize bacterial, fungal, and other microbial carbohydrates prevent systemic infections and help maintain microbiome homeostasis. Anti-glycan antibodies can have both beneficial and detrimental effects. For example, alloantibodies to ABO blood group carbohydrates can help reduce the spread of some infectious diseases, but they also impose limitations for blood transfusions. Antibodies that recognize self-glycans can contribute to autoimmune diseases, such as Guillain-Barre syndrome. In addition to endogenous antibodies that arise through natural processes, a variety of vaccines induce anti-glycan antibodies as a primary mechanism of protection. Some examples of approved carbohydrate-based vaccines that have had a major impact on human health are against pneumococcus, Haemophilus influeanza type b, and Neisseria meningitidis. Monoclonal antibodies specifically targeting pathogen associated or tumor associated carbohydrate antigens (TACAs) are used clinically for both diagnostic and therapeutic purposes. This review aims to highlight some of the well-studied and critically important applications of anti-carbohydrate antibodies.


Assuntos
Síndrome de Guillain-Barré/imunologia , Infecções por Haemophilus/imunologia , Meningite Meningocócica/imunologia , Pneumonia Pneumocócica/imunologia , Polissacarídeos/imunologia , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/uso terapêutico , Autoanticorpos/biossíntese , Autoanticorpos/sangue , Vacinas Bacterianas/biossíntese , Vacinas Bacterianas/uso terapêutico , Sequência de Carboidratos , Síndrome de Guillain-Barré/patologia , Infecções por Haemophilus/microbiologia , Infecções por Haemophilus/prevenção & controle , Vacinas Anti-Haemophilus/biossíntese , Vacinas Anti-Haemophilus/uso terapêutico , Haemophilus influenzae/imunologia , Humanos , Meningite Meningocócica/microbiologia , Meningite Meningocócica/prevenção & controle , Neisseria meningitidis/imunologia , Vacinas Pneumocócicas/biossíntese , Vacinas Pneumocócicas/uso terapêutico , Pneumonia Pneumocócica/microbiologia , Pneumonia Pneumocócica/prevenção & controle , Polissacarídeos/antagonistas & inibidores , Polissacarídeos/química , Streptococcus pneumoniae/imunologia
8.
Am J Respir Cell Mol Biol ; 64(5): 579-591, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33625952

RESUMO

Community-acquired pneumonia is the most common type of pneumonia and remains a leading cause of morbidity and mortality worldwide. Although many different pathogens can contribute to pneumonia, Streptococcus pneumoniae is one of the common bacterial pathogens that underlie community-acquired pneumonia. RIPK3 (receptor-interacting protein kinase 3) is widely recognized as a key modulator of inflammation and cell death. To elucidate a potential role of RIPK3 in pneumonia, we examined plasma from healthy control subjects and patients positive for streptococcal pneumonia. In human studies, RIPK3 protein concentrations were significantly elevated and were identified as a potential plasma marker of pneumococcal pneumonia. To expand these findings, we used an in vivo murine model of pneumococcal pneumonia to demonstrate that RIPK3 deficiency leads to reduced bacterial clearance, severe pathological damage, and high mortality. Our results illustrated that RIPK3 forms a complex with RIPK1, MLKL (mixed-lineage kinase domain-like protein), and MCU (mitochondrial calcium uniporter) to induce mitochondrial calcium uptake and mitochondrial reactive oxygen species(mROS) production during S. pneumoniae infection. In macrophages, RIPK3 initiated necroptosis via the mROS-mediated mitochondrial permeability transition pore opening and NLRP3 inflammasome activation via the mROS-AKT pathway to protect against S. pneumoniae. In conclusion, our study demonstrated a mechanism by which RIPK3-initiated necroptosis is essential for host defense against S. pneumoniae.


Assuntos
Macrófagos Alveolares/imunologia , Mitocôndrias/imunologia , Pneumonia Pneumocócica/imunologia , Proteínas Quinases/imunologia , Proteína Serina-Treonina Quinases de Interação com Receptores/imunologia , Streptococcus pneumoniae/patogenicidade , Idoso , Animais , Canais de Cálcio/genética , Canais de Cálcio/imunologia , Estudos de Casos e Controles , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Humanos , Inflamassomos/genética , Inflamassomos/imunologia , Macrófagos Alveolares/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Mitocôndrias/patologia , Poro de Transição de Permeabilidade Mitocondrial/imunologia , Poro de Transição de Permeabilidade Mitocondrial/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Necroptose/genética , Necroptose/imunologia , Pneumonia Pneumocócica/complicações , Pneumonia Pneumocócica/microbiologia , Proteínas Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/imunologia , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Transdução de Sinais , Streptococcus pneumoniae/imunologia
9.
Nutrients ; 13(1)2021 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-33478006

RESUMO

Vitamin D is an essential component of immune function and childhood deficiency is associated with an increased risk of acute lower respiratory infections (ALRIs). Globally, the leading childhood respiratory pathogens are Streptococcus pneumoniae, respiratory syncytial virus and the influenza virus. There is a growing body of evidence describing the innate immunomodulatory properties of vitamin D during challenge with respiratory pathogens, but recent systematic and unbiased synthesis of data is lacking, and future research directions are unclear. We therefore conducted a systematic PubMed literature search using the terms "vitamin D" and "Streptococcus pneumoniae" or "Respiratory Syncytial Virus" or "Influenza". A priori inclusion criteria restricted the review to in vitro studies investigating the effect of vitamin D metabolites on human innate immune cells (primary, differentiated or immortalised) in response to stimulation with the specified respiratory pathogens. Eleven studies met our criteria. Despite some heterogeneity across pathogens and innate cell types, vitamin D modulated pathogen recognition receptor (PRRs: Toll-like receptor 2 (TLR2), TLR4, TLR7 and nucleotide-binding oligomerisation domain-containing protein 2 (NOD2)) expression; increased antimicrobial peptide expression (LL-37, human neutrophil peptide (HNP) 1-3 and ß-defensin); modulated autophagosome production reducing apoptosis; and modulated production of inflammatory cytokines (Interleukin (IL) -1ß, tumour necrosis factor-α (TNF-α), interferon-É£ (IFN-É£), IL-12p70, IFN-ß, Regulated on Activation, Normal T cell Expressed (RANTES), IL-10) and chemokines (IL-8 and C-X-C motif chemokine ligand 10 (CXCL10)). Differential modulation of PRRs and IL-1ß was reported across immune cell types; however, this may be due to the experimental design. None of the studies specifically focused on immune responses in cells derived from children. In summary, vitamin D promotes a balanced immune response, potentially enhancing pathogen sensing and clearance and restricting pathogen induced inflammatory dysregulation. This is likely to be important in controlling both ALRIs and the immunopathology associated with poorer outcomes and progression to chronic lung diseases. Many unknowns remain and further investigation is required to clarify the nuances in vitamin D mediated immune responses by pathogen and immune cell type and to determine whether these in vitro findings translate into enhanced immunity and reduced ALRI in the paediatric clinical setting.


Assuntos
Imunidade Inata , Vírus da Influenza A/imunologia , Vírus Sinciciais Respiratórios/imunologia , Infecções Respiratórias/imunologia , Streptococcus pneumoniae/imunologia , Vitamina D/metabolismo , Vitamina D/farmacologia , Criança , Pré-Escolar , Citocinas/metabolismo , Humanos , Imunomodulação , Lactente , Influenza Humana/imunologia , Influenza Humana/virologia , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/microbiologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Infecções Respiratórias/microbiologia , Infecções Respiratórias/virologia
10.
J Mol Biol ; 433(2): 166723, 2021 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-33242497

RESUMO

Nucleotides are important for RNA and DNA synthesis and, despite a de novo synthesis by bacteria, uptake systems are crucial. Streptococcus pneumoniae, a facultative human pathogen, produces a surface-exposed nucleoside-binding protein, PnrA, as part of an ABC transporter system. Here we demonstrate the binding affinity of PnrA to nucleosides adenosine, guanosine, cytidine, thymidine and uridine by microscale thermophoresis and indicate the consumption of adenosine and guanosine by 1H NMR spectroscopy. In a series of five crystal structures we revealed the PnrA structure and provide insights into how PnrA can bind purine and pyrimidine ribonucleosides but with preference for purine ribonucleosides. Crystal structures of PnrA:nucleoside complexes unveil a clear pattern of interactions in which both the N- and C- domains of PnrA contribute. The ribose moiety is strongly recognized through a conserved network of H-bond interactions, while plasticity in loop 27-36 is essential to bind purine- or pyrimidine-based nucleosides. Further, we deciphered the role of PnrA in pneumococcal fitness in infection experiments. Phagocytosis experiments did not show a clear difference in phagocytosis between PnrA-deficient and wild-type pneumococci. In the acute pneumonia infection model the deficiency of PnrA attenuated moderately virulence of the mutant, which is indicated by a delay in the development of severe lung infections. Importantly, we confirmed the loss of fitness in co-infections, where the wild-type out-competed the pnrA-mutant. In conclusion, we present the PnrA structure in complex with individual nucleosides and show that the consumption of adenosine and guanosine under infection conditions is required for virulence.


Assuntos
Transportadores de Cassetes de Ligação de ATP/química , Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Modelos Moleculares , Streptococcus pneumoniae/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Proteínas de Bactérias/genética , Cristalografia por Raios X , Modelos Animais de Doenças , Humanos , Ligação de Hidrogênio , Cinética , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Nucleosídeos/química , Nucleosídeos/metabolismo , Fagocitose , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/metabolismo , Pneumonia Pneumocócica/microbiologia , Pneumonia Pneumocócica/patologia , Ligação Proteica , Conformação Proteica , Streptococcus pneumoniae/imunologia , Relação Estrutura-Atividade
11.
JCI Insight ; 6(3)2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33290273

RESUMO

Current treatments for pneumonia (PNA) are focused on the pathogens. Mortality from PNA-induced acute lung injury (PNA-ALI) remains high, underscoring the need for additional therapeutic targets. Clinical and experimental evidence exists for potential sex differences in PNA survival, with males having higher mortality. In a model of severe pneumococcal PNA, when compared with male mice, age-matched female mice exhibited enhanced resolution characterized by decreased alveolar and lung inflammation and increased numbers of Tregs. Recognizing the critical role of Tregs in lung injury resolution, we evaluated whether improved outcomes in female mice were due to estradiol (E2) effects on Treg biology. E2 promoted a Treg-suppressive phenotype in vitro and resolution of PNA in vivo. Systemic rescue administration of E2 promoted resolution of PNA in male mice independent of lung bacterial clearance. E2 augmented Treg expression of Foxp3, CD25, and GATA3, an effect that required ERß, and not ERα, signaling. Importantly, the in vivo therapeutic effects of E2 were lost in Treg-depleted mice (Foxp3DTR mice). Adoptive transfer of ex vivo E2-treated Tregs rescued Streptococcus pneumoniae-induce PNA-ALI, a salutary effect that required Treg ERß expression. E2/ERß was required for Tregs to control macrophage proinflammatory responses. Our findings support the therapeutic role for E2 in promoting resolution of lung inflammation after PNA via ERß Tregs.


Assuntos
Estradiol/farmacologia , Receptor beta de Estrogênio/metabolismo , Pneumonia Pneumocócica/tratamento farmacológico , Linfócitos T Reguladores/efeitos dos fármacos , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/metabolismo , Transferência Adotiva , Animais , Modelos Animais de Doenças , Estradiol/metabolismo , Receptor beta de Estrogênio/deficiência , Receptor beta de Estrogênio/genética , Feminino , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/metabolismo , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/patologia , Fatores Sexuais , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
12.
J Immunol ; 205(2): 469-479, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32540994

RESUMO

Allergic asthma (AA) is characterized as a Th2-biased airway inflammation that can develop lung inflammation and remodeling of the respiratory tract. Streptococcus pneumoniae is a major respiratory pathogen, causing noninvasive (otitis media and pneumonia) and invasive diseases (sepsis) in humans. We sought to determine the role of IL-6 in the regulation of lung inflammation in murine AA caused by Aspergillus fumigatus as well as its consequence on the regulation of airway barrier integrity and S. pneumoniae disease. In an AA model, IL-6 deficiency led to increased lung inflammation, eosinophil recruitment, tissue pathology, and collagen deposition. Additionally, IL-6-deficient asthmatic mice exhibited reduced goblet cell hyperplasia and increased TGF-ß production. These key changes in the lungs of IL-6-deficient asthmatic mice resulted in dysregulated tight junction proteins and increased lung permeability. Whereas the host response to AA protected against S. pneumoniae lung disease, the IL-6 deficiency abrogated the protective effect of allergic inflammation against S. pneumoniae pathogenesis. Consistent with in vivo data, IL-6 knockdown by small interfering RNA or the blockade of IL-6R signaling exacerbated the TGF-ß-induced dysregulation of tight junction proteins, E-cadherin and N-cadherin expression, and STAT3 phosphorylation in MLE-12 epithelial cells. Our findings demonstrate a previously unrecognized role of host IL-6 response in the regulation of lung inflammation during AA and the control of S. pneumoniae bacterial disease. A better understanding of the interactions between lung inflammation and barrier framework could lead to the development of therapies to control asthma inflammation and preserve barrier integrity.


Assuntos
Asma/imunologia , Células Caliciformes/patologia , Hipersensibilidade/imunologia , Interleucina-6/metabolismo , Pneumonia Pneumocócica/imunologia , Pneumonia/imunologia , Mucosa Respiratória/metabolismo , Streptococcus pneumoniae/fisiologia , Junções Íntimas/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Resistência à Doença , Humanos , Hiperplasia , Interleucina-6/genética , Camundongos , Camundongos Knockout , RNA Interferente Pequeno/genética , Mucosa Respiratória/patologia , Transdução de Sinais , Fator de Crescimento Transformador beta/sangue
13.
J Clin Invest ; 130(6): 3021-3037, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32364537

RESUMO

Patients with respiratory syncytial virus (RSV) infection exhibit enhanced susceptibility to subsequent pneumococcal infections. However, the underlying mechanisms involved in this increased susceptibility remain unclear. Here, we identified potentially novel cellular and molecular cascades triggered by RSV infection to exacerbate secondary pneumococcal pneumonia. RSV infection stimulated the local production of growth arrest-specific 6 (Gas6). The Gas6 receptor Axl was crucial for attenuating pneumococcal immunity in that the Gas6/Axl blockade fully restored antibacterial immunity. Mechanistically, Gas6/Axl interaction regulated the conversion of alveolar macrophages from an antibacterial phenotype to an M2-like phenotype that did not exhibit antibacterial activity, and the attenuation of caspase-1 activation and IL-18 production in response to pneumococcal infection. The attenuated IL-18 production failed to drive both NK cell-mediated IFN-γ production and local NO and TNF-α production, which impair the control of bacterial infection. Hence, the RSV-mediated Gas6/Axl activity attenuates the macrophage-mediated protection against pneumococcal infection. The Gas6/Axl axis could be a potentially novel therapeutic target for RSV-associated secondary bacterial infection.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Macrófagos Alveolares/imunologia , Pneumonia Pneumocócica/imunologia , Proteínas Proto-Oncogênicas/imunologia , Receptores Proteína Tirosina Quinases/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Animais , Citocinas/genética , Citocinas/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Macrófagos Alveolares/microbiologia , Macrófagos Alveolares/patologia , Macrófagos Alveolares/virologia , Masculino , Camundongos , Camundongos Knockout , Pneumonia Pneumocócica/genética , Pneumonia Pneumocócica/patologia , Pneumonia Pneumocócica/virologia , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Infecções por Vírus Respiratório Sincicial/genética , Infecções por Vírus Respiratório Sincicial/patologia , Receptor Tirosina Quinase Axl
14.
Front Immunol ; 11: 554, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32300347

RESUMO

Cyclic di-AMP (c-di-AMP) is an important signaling molecule for pneumococci, and as a uniquely prokaryotic product it can be recognized by mammalian cells as a danger signal that triggers innate immunity. Roles of c-di-AMP in directing host responses during pneumococcal infection are only beginning to be defined. We hypothesized that pneumococci with defective c-di-AMP catabolism due to phosphodiesterase deletions could illuminate roles of c-di-AMP in mediating host responses to pneumococcal infection. Pneumococci deficient in phosphodiesterase 2 (Pde2) stimulated a rapid induction of interferon ß (IFNß) expression that was exaggerated in comparison to that induced by wild type (WT) bacteria or bacteria deficient in phosphodiesterase 1. This IFNß burst was elicited in mouse and human macrophage-like cell lines as well as in primary alveolar macrophages collected from mice with pneumococcal pneumonia. Macrophage hyperactivation by Pde2-deficient pneumococci led to rapid cell death. STING and cGAS were essential for the excessive IFNß induction, which also required phagocytosis of bacteria and triggered the phosphorylation of IRF3 and IRF7 transcription factors. The select effects of Pde2 deletion were products of a unique role of this enzyme in c-di-AMP catabolism when pneumococci were grown on solid substrate conditions designed to enhance virulence. Because pneumococci with elevated c-di-AMP drive aberrant innate immune responses from macrophages involving hyperactivation of STING, excessive IFNß expression, and rapid cytotoxicity, we surmise that c-di-AMP is pivotal for directing innate immunity and host-pathogen interactions during pneumococcal pneumonia.


Assuntos
Proteínas de Bactérias/imunologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/imunologia , Fosfatos de Dinucleosídeos/imunologia , Imunidade Inata/imunologia , Macrófagos/imunologia , Streptococcus pneumoniae/imunologia , Animais , Proteínas de Bactérias/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/metabolismo , Fosfatos de Dinucleosídeos/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia Pneumocócica/imunologia , Células RAW 264.7
15.
Physiol Rep ; 8(3): e14368, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32061190

RESUMO

The immunologic responses that occur early in the acute respiratory distress syndrome (ARDS) elicit immune-mediated damage. The mechanisms underlying the resolution of ARDS, particularly the role of signaling molecules in regulating immune cell kinetics, remain important questions. Th1-mediated responses can contribute to the pathogenesis of acute lung injury (ALI). Interferon-gamma (IFN-γ) orchestrates early inflammatory events, enhancing immune-mediated damage. The current study investigated IFN-γ during resolution in several experimental models of ALI. The absence of IFN-γ resulted in altered kinetics of lymphocyte and macrophage responses, suggesting that IFN-γ present in this microenvironment is influential in ALI resolution. Genetic deficiency of IFN-γ or administering neutralizing IFN-γ antibodies accelerated the pace of resolution. Neutralizing IFN-γ decreased the numbers of interstitial and inflammatory macrophages and increased alveolar macrophage numbers during resolution. Our results underline the complexity of lung injury resolution and provide insight into the effects through which altered IFN-γ concentrations affect immune cell kinetics and the rate of resolution. These findings suggest that therapies that spatially or temporally control IFN-γ signaling may promote ALI resolution. Identifying and elucidating the mechanisms critical to ALI resolution will allow the development of therapeutic approaches to minimize collateral tissue damage without adversely altering the response to injury.


Assuntos
Interferon gama/metabolismo , Pneumonia Pneumocócica/imunologia , Síndrome do Desconforto Respiratório/imunologia , Animais , Feminino , Interferon gama/genética , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Reguladores/imunologia
16.
J Leukoc Biol ; 108(3): 867-882, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32017200

RESUMO

Polymorphonuclear leukocytes (PMNs) are crucial for initial control of Streptococcus pneumoniae (pneumococcus) lung infection; however, as the infection progresses their persistence in the lungs becomes detrimental. Here we explored why the antimicrobial efficacy of PMNs declines over the course of infection. We found that the progressive inability of PMNs to control infection correlated with phenotypic differences characterized by a decrease in CD73 expression, an enzyme required for production of extracellular adenosine (EAD). EAD production by CD73 was crucial for the ability of both murine and human PMNs to kill S. pneumoniae. In exploring the mechanisms by which CD73 controlled PMN function, we found that CD73 mediated its antimicrobial activity by inhibiting IL-10 production. PMNs from wild-type mice did not increase IL-10 production in response to S. pneumoniae; however, CD73-/- PMNs up-regulated IL-10 production upon pneumococcal infection in vitro and during lung challenge. IL-10 inhibited the ability of WT PMNs to kill pneumococci. Conversely, blocking IL-10 boosted the bactericidal activity of CD73-/- PMNs as well as host resistance of CD73-/- mice to pneumococcal pneumonia. CD73/IL-10 did not affect apoptosis, bacterial uptake, and intracellular killing or production of antimicrobial neutrophil elastase and myeloperoxidase. Rather, inhibition of IL-10 production by CD73 was important for optimal reactive oxygen species (ROS) production by PMNs. ROS contributed to PMN antimicrobial function as their removal or detoxification impaired the ability of PMNs to efficiently kill S. pneumoniae. This study demonstrates that CD73 controls PMN antimicrobial phenotype during S. pneumoniae infection.


Assuntos
5'-Nucleotidase/fisiologia , Adenosina/fisiologia , Interleucina-10/biossíntese , Neutrófilos/enzimologia , Pneumonia Pneumocócica/imunologia , Streptococcus pneumoniae , 5'-Nucleotidase/biossíntese , 5'-Nucleotidase/deficiência , 5'-Nucleotidase/genética , Adenosina/biossíntese , Transferência Adotiva , Adulto , Animais , Proteínas de Bactérias/genética , Grânulos Citoplasmáticos/enzimologia , Regulação para Baixo , Indução Enzimática , Líquido Extracelular , Feminino , Proteínas Ligadas por GPI/fisiologia , Humanos , Interleucina-10/genética , Elastase de Leucócito/biossíntese , Elastase de Leucócito/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/fisiologia , Peroxidase/biossíntese , Peroxidase/genética , Pneumonia Pneumocócica/prevenção & controle , Espécies Reativas de Oxigênio/metabolismo , Streptococcus pneumoniae/enzimologia , Streptococcus pneumoniae/genética , Superóxido Dismutase/deficiência , Superóxido Dismutase/genética , Adulto Jovem
17.
Microbes Infect ; 22(8): 312-321, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-31958572

RESUMO

Pneumococcal conjugate vaccination (PCV) may prevent influenza-related pneumonia, including Streptococcus pneumoniae pneumonia. To investigate PCV efficacy against secondary pneumococcal pneumonia following influenza, PCV was administered intramuscularly 2 and 5 weeks before S. pneumoniae serotype-3 colonization of murine nasopharynges followed by intranasal challenge with a sublethal dose of influenza A virus. Bacterial and viral loads, including innate immune responses were compared across conditions. PCV vaccination improved the survival of mice with secondary pneumococcal pneumonia and significantly reduced the pulmonary bacterial burden. Increased monocyte/macrophage influx into the lungs, alleviated loss of alveolar macrophages and decreased neutrophil influx into the lungs occurred in PCV-treated mice irrespective of pneumococcal colonization. Higher monocyte chemoattractant protein 1 levels and lower levels of CXCL1, interferon-γ, interleukin-17A, and IL-10, were detected in PCV-treated mice. Additionally, PCV treatment activated the macrophage intracellular killing of S. pneumoniae. Collectively, PCV potentially modulates the host's innate immunity and specific antibodies induction. Macrophage-related innate immunity should be further explored to elucidate the efficacy and mechanisms of PCV versus influenza-related life-threatening diseases.


Assuntos
Coinfecção/imunologia , Imunidade Inata , Macrófagos/imunologia , Infecções por Orthomyxoviridae/imunologia , Vacinas Pneumocócicas/imunologia , Pneumonia Pneumocócica/imunologia , Animais , Anticorpos Antibacterianos/sangue , Antígeno B7-2/metabolismo , Carga Bacteriana , Coinfecção/microbiologia , Coinfecção/mortalidade , Coinfecção/virologia , Citocinas/metabolismo , Modelos Animais de Doenças , Vírus da Influenza A , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/virologia , Macrófagos/microbiologia , Camundongos , Neutrófilos/imunologia , Infecções por Orthomyxoviridae/microbiologia , Infecções por Orthomyxoviridae/mortalidade , Infecções por Orthomyxoviridae/virologia , Fagocitose , Vacinas Pneumocócicas/administração & dosagem , Pneumonia Pneumocócica/microbiologia , Pneumonia Pneumocócica/mortalidade , Pneumonia Pneumocócica/virologia , Streptococcus pneumoniae , Taxa de Sobrevida , Vacinação , Vacinas Conjugadas/administração & dosagem , Vacinas Conjugadas/imunologia
18.
J Allergy Clin Immunol ; 145(4): 1272-1284.e6, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31983527

RESUMO

BACKGROUND: The World Health Organization estimates that air pollution is responsible for 7 million deaths per annum, with 7% of these attributable to pneumonia. Many of these fatalities have been linked to exposure to high levels of airborne particulates, such as diesel exhaust particles (DEPs). OBJECTIVES: We sought to determine whether exposure to DEPs could promote the progression of asymptomatic nasopharyngeal carriage of Streptococcus pneumoniae to invasive pneumococcal disease. METHODS: We used mouse models and in vitro assays to provide a mechanistic understanding of the link between DEP exposure and pneumococcal disease risk, and we confirmed our findings by using induced sputum macrophages isolated from healthy human volunteers. RESULTS: We demonstrate that inhaled exposure to DEPs disrupts asymptomatic nasopharyngeal carriage of S pneumoniae in mice, leading to dissemination to lungs and blood. Pneumococci are transported from the nasopharynx to the lungs following exposure to DEPs, leading to increased proinflammatory cytokine production, reduced phagocytic function of alveolar macrophages, and consequently, increased pneumococcal loads within the lungs and translocation into blood. These findings were confirmed by using DEP-exposed induced sputum macrophages isolated from healthy volunteers, demonstrating that impaired innate immune mechanisms following DEP exposure are also at play in humans. CONCLUSION: Lung inhaled DEPs increase susceptibility to pneumococcal disease by leading to loss of immunological control of pneumococcal colonisation, increased inflammation, tissue damage, and systemic bacterial dissemination.


Assuntos
Pulmão/imunologia , Macrófagos/imunologia , Nasofaringe/patologia , Material Particulado/efeitos adversos , Pneumonia Pneumocócica/imunologia , Streptococcus pneumoniae/fisiologia , Animais , Bacteriemia , Portador Sadio , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Suscetibilidade a Doenças , Humanos , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nasofaringe/microbiologia , Fagocitose , Pneumonia Pneumocócica/epidemiologia , Risco , Emissões de Veículos
19.
Front Immunol ; 11: 611661, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33584691

RESUMO

The anti-CD20 antibody Rituximab to deplete CD20+ B cells is an effective treatment for rheumatoid arthritis and B cell malignancies, but is associated with an increased incidence of respiratory infections. Using mouse models we have investigated the consequences of B cell depletion on natural and acquired humoral immunity to Streptococcus pneumoniae. B cell depletion of naïve C57Bl/6 mice reduced natural IgM recognition of S. pneumoniae, but did not increase susceptibility to S. pneumoniae pneumonia. ELISA and flow cytometry assays demonstrated significantly reduced IgG and IgM recognition of S. pneumoniae in sera from mice treated with B cell depletion prior to S. pneumoniae nasopharyngeal colonization compared to untreated mice. Colonization induced antibody responses to protein rather than capsular antigen, and when measured using a protein array B cell depletion prior to colonization reduced serum levels of IgG to several protein antigens. However, B cell depleted S. pneumoniae colonized mice were still partially protected against both lung infection and septicemia when challenged with S. pneumoniae after reconstitution of their B cells. These data indicate that although B cell depletion markedly impairs antibody recognition of S. pneumoniae in colonized mice, some protective immunity is maintained, perhaps mediated by cellular immunity.


Assuntos
Linfócitos B/efeitos dos fármacos , Imunidade Humoral , Imunidade Inata , Fatores Imunológicos/farmacologia , Depleção Linfocítica , Pneumonia Pneumocócica/prevenção & controle , Rituximab/farmacologia , Streptococcus pneumoniae/imunologia , Animais , Anticorpos Antibacterianos/sangue , Linfócitos B/imunologia , Modelos Animais de Doenças , Feminino , Interações Hospedeiro-Patógeno , Imunidade Celular , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Camundongos Endogâmicos C57BL , Pneumonia Pneumocócica/sangue , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/microbiologia , Streptococcus pneumoniae/patogenicidade
20.
Cell Microbiol ; 22(2): e13141, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31709673

RESUMO

Extracellular adenosine production is crucial for host resistance against Streptococcus pneumoniae (pneumococcus) and is thought to affect antibacterial immune responses by neutrophils. However, whether extracellular adenosine alters direct host-pathogen interaction remains unexplored. An important determinant for lung infection by S. pneumoniae is its ability to adhere to the pulmonary epithelium. Here we explored whether extracellular adenosine can directly impact bacterial adherence to lung epithelial cells. We found that signaling via A1 adenosine receptor significantly reduced the ability of pneumococci to bind human pulmonary epithelial cells. A1 receptor signaling blocked bacterial binding by reducing the expression of platelet-activating factor receptor, a host protein used by S. pneumoniae to adhere to host cells. In vivo, A1 was required for control of pneumococcal pneumonia as inhibiting it resulted in increased host susceptibility. As S. pneumoniae remain a leading cause of community-acquired pneumonia in the elderly, we explored the role of A1 in the age-driven susceptibility to infection. We found no difference in A1 pulmonary expression in young versus old mice. Strikingly, triggering A1 signaling boosted host resistance of old mice to S. pneumoniae pulmonary infection. This study demonstrates a novel mechanism by which extracellular adenosine modulates resistance to lung infection by targeting bacterial-host interactions.


Assuntos
Células Epiteliais/microbiologia , Glicoproteínas da Membrana de Plaquetas/metabolismo , Pneumonia Pneumocócica , Receptor A1 de Adenosina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Streptococcus pneumoniae , Fatores Etários , Animais , Aderência Bacteriana , Linhagem Celular , Células Epiteliais/citologia , Células Epiteliais/imunologia , Interações Hospedeiro-Patógeno , Humanos , Pulmão/citologia , Pulmão/imunologia , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/microbiologia , Streptococcus pneumoniae/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA