Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Immunol Cell Biol ; 101(2): 130-141, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36318273

RESUMO

Protein kinase D (PKD) is a serine/threonine kinase family with three isoforms (PKD1-3) that are expressed in most cells and implicated in a wide array of signaling pathways, including cell growth, differentiation, transcription, secretion, polarization and actin turnover. Despite growing interest in PKD, relatively little is known about the role of PKD in immune responses. We recently published that inhibiting PKD limits proinflammatory cytokine secretion and leukocyte accumulation in mouse models of viral infection, and that PKD3 is highly expressed in the murine lung and immune cell populations. Here we focus on the immune-related phenotypes of PKD3 knockout mice. We report that PKD3 is necessary for maximal neutrophil accumulation in the lung following challenge with inhaled polyinosinic:polycytidylic acid, a double-stranded RNA, as well as following influenza A virus infection. Using reciprocal bone marrow chimeras, we found that PKD3 is required in the hematopoietic compartment for optimal neutrophil migration to the lung. Ex vivo transwell and chemokinesis assays confirmed that PKD3-/- neutrophils possess an intrinsic motility defect, partly because of reduced surface expression of CD18, which is critical for leukocyte migration. Finally, the peak of neutrophilia was significantly reduced in PKD3-/- mice after lethal influenza A virus infection. Together, these results demonstrate that PKD3 has an essential, and nonredundant, role in promoting neutrophil recruitment to the lung. A better understanding of the isoform-specific and cell type-specific activities of PKD has the potential to lead to novel therapeutics for respiratory illnesses.


Assuntos
Neutrófilos , Proteína Quinase C , Viroses , Animais , Camundongos , Neutrófilos/metabolismo , Isoformas de Proteínas , Transdução de Sinais , Proteína Quinase C/metabolismo
2.
Part Fibre Toxicol ; 17(1): 52, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-33059747

RESUMO

BACKGROUND: While exposure to diesel exhaust particles has been linked to aberrant immune responses in allergic diseases such as asthma, little attention has been paid to their effects on the airway epithelial barrier. In this study, we sought to determine the effect of diesel exhaust exposure on airway epithelial barrier function and composition using in vitro and in vivo model systems. METHODS: 16HBE14o- human bronchial epithelial cells were grown on collagen coated Transwell inserts and exposed to 5 to 50 µg/cm2 SRM 2975 diesel particulate matter (DEP) suspended in cell culture medium or vehicle controls. Changes in barrier function were assessed by measuring transepithelial electrical resistance (TEER) and permeability to 4 kDa FITC Dextran. Neonatal BALB/c mice were exposed to aerosolized DEP (255 ± 89 µg/m3; 2 h per day for 5 days) and changes in the tight junction protein Tricellulin were assessed 2 weeks post exposure. RESULTS: A six-hour incubation of epithelial cells with diesel exhaust particles caused a significant concentration-dependent reduction in epithelial barrier integrity as measured by decreased TEER and increased permeability to 4 kDa FITC-Dextran. This reduction in epithelial barrier integrity corresponded to a significant reduction in expression of the tight junction protein Tricellulin. siRNA mediated knockdown of Tricellulin recapitulated changes in barrier function caused by DEP exposure. Neonatal exposure to aerosolized DEP caused a significant reduction in lung Tricellulin 2 weeks post exposure at both the protein and mRNA level. CONCLUSION: Short term exposure to DEP causes a significant reduction in epithelial barrier integrity through a reduction in the tight junction protein Tricellulin. Neonatal exposure to aerosolized DEP caused a significant and sustained reduction in Tricellulin protein and mRNA in the lung, suggesting that early life exposure to inhaled DEP may cause lasting changes in airway epithelial barrier function.


Assuntos
Poluentes Atmosféricos/toxicidade , Proteína 2 com Domínio MARVEL/metabolismo , Emissões de Veículos/toxicidade , Animais , Asma , Células Epiteliais , Humanos , Pulmão , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Junções Íntimas
3.
J Asthma ; 56(12): 1257-1265, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-30444143

RESUMO

Objective: Airway epithelial barrier dysfunction is emerging as an important feature of asthma pathogenesis, but this is difficult to measure in individual subjects. We aimed to develop a noninvasive way to measure airway permeability in asthma. Methods: Healthy controls and subjects with mild asthma inhaled dry powder mannitol in a dose-escalating manner on two separate occasions, stopping at 155 mg or 315 mg. Serum mannitol levels were measured at baseline and then 30, 90, and 150 min after mannitol inhalation. Mannitol absorption was compared with measurements of airflow obstruction (FEV1) and airway inflammation (FeNO). Results: Serum mannitol levels increased in a time- and dose-dependent manner in both healthy control and subjects with asthma. There were no significant differences in mannitol absorption when comparing healthy controls and subjects with asthma. Mannitol absorption did not correlate with markers of airway obstruction or inflammation. Conclusions: Measuring serum concentrations of mannitol after inhalation challenge can potentially provide insights into airway barrier function in asthma.


Assuntos
Antiasmáticos/administração & dosagem , Asma/diagnóstico , Epitélio/patologia , Volume Expiratório Forçado/efeitos dos fármacos , Manitol/administração & dosagem , Manitol/sangue , Administração por Inalação , Manuseio das Vias Aéreas , Remodelação das Vias Aéreas/efeitos dos fármacos , Análise de Variância , Área Sob a Curva , Asma/tratamento farmacológico , Testes de Provocação Brônquica/métodos , Estudos de Casos e Controles , Relação Dose-Resposta a Droga , Esquema de Medicação , Epitélio/efeitos dos fármacos , Feminino , Humanos , Pulmão/efeitos dos fármacos , Pulmão/fisiopatologia , Masculino , Projetos Piloto , Valores de Referência , Índice de Gravidade de Doença , Estatísticas não Paramétricas , Fatores de Tempo
4.
PLoS One ; 19(7): e0306666, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38950013

RESUMO

[This corrects the article DOI: 10.1371/journal.pone.0260706.].

5.
Sci Immunol ; 9(92): eadf8776, 2024 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-38394230

RESUMO

CD8+ T cells are classically recognized as adaptive lymphocytes based on their ability to recognize specific foreign antigens and mount memory responses. However, recent studies indicate that some antigen-inexperienced CD8+ T cells can respond to innate cytokines alone in the absence of cognate T cell receptor stimulation, a phenomenon referred to as bystander activation. Here, we demonstrate that neonatal CD8+ T cells undergo a robust and diverse program of bystander activation, which corresponds to enhanced innate-like protection against unrelated pathogens. Using a multi-omics approach, we found that the ability of neonatal CD8+ T cells to respond to innate cytokines derives from their capacity to undergo rapid chromatin remodeling, resulting in the usage of a distinct set of enhancers and transcription factors typically found in innate-like T cells. We observed that the switch between innate and adaptive functions in the CD8+ T cell compartment is mediated by changes in the abundance of distinct subsets of cells. The innate CD8+ T cell subset that predominates in early life was also present in adult mice and humans. Our findings provide support for the layered immune hypothesis and indicate that the CD8+ T cell compartment is more functionally diverse than previously thought.


Assuntos
Linfócitos T CD8-Positivos , Imunidade Inata , Humanos , Adulto , Camundongos , Animais , Citocinas , Subpopulações de Linfócitos T , Antígenos
6.
PLoS One ; 16(12): e0260706, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34871316

RESUMO

Airway epithelial barrier dysfunction is increasingly recognized as a key feature of asthma and other lung diseases. Respiratory viruses are responsible for a large fraction of asthma exacerbations, and are particularly potent at disrupting epithelial barrier function through pattern recognition receptor engagement leading to tight junction dysfunction. Although different mechanisms of barrier dysfunction have been described, relatively little is known about whether barrier integrity can be promoted to limit disease. Here, we tested three classes of drugs commonly prescribed to treat asthma for their ability to promote barrier function using a cell culture model of virus-induced airway epithelial barrier disruption. Specifically, we studied the corticosteroid budesonide, the long acting beta-agonist formoterol, and the leukotriene receptor antagonist montelukast for their ability to promote barrier integrity of a monolayer of human bronchial epithelial cells (16HBE) before exposure to the viral mimetic double-stranded RNA. Of the three, only budesonide treatment limited transepithelial electrical resistance and small molecule permeability (4 kDa FITC-dextran flux). Next, we used a mouse model of acute dsRNA challenge that induces transient epithelial barrier disruption in vivo, and studied the effects budesonide when administered prophylactically or therapeutically. We found that budesonide similarly protected against dsRNA-induced airway barrier disruption in the lung, independently of its effects on airway inflammation. Taken together, these data suggest that an under-appreciated effect of inhaled budesonide is to maintain or promote airway epithelial barrier integrity during respiratory viral infections.


Assuntos
Asma/tratamento farmacológico , Brônquios/efeitos dos fármacos , Broncodilatadores/farmacologia , Budesonida/farmacologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Poli I-C/antagonistas & inibidores , Acetatos/farmacologia , Administração por Inalação , Animais , Asma/induzido quimicamente , Asma/metabolismo , Asma/patologia , Brônquios/metabolismo , Brônquios/patologia , Linhagem Celular , Ciclopropanos/farmacologia , Dextranos/metabolismo , Impedância Elétrica , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Fumarato de Formoterol/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Mimetismo Molecular , Poli I-C/farmacologia , Quinolinas/farmacologia , RNA de Cadeia Dupla/antagonistas & inibidores , RNA de Cadeia Dupla/farmacologia , RNA Viral/antagonistas & inibidores , RNA Viral/farmacologia , Sulfetos/farmacologia , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo
7.
J Clin Invest ; 131(1)2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33079726

RESUMO

Although platelets are the cellular mediators of thrombosis, they are also immune cells. Platelets interact both directly and indirectly with immune cells, impacting their activation and differentiation, as well as all phases of the immune response. Megakaryocytes (Mks) are the cell source of circulating platelets, and until recently Mks were typically only considered bone marrow-resident (BM-resident) cells. However, platelet-producing Mks also reside in the lung, and lung Mks express greater levels of immune molecules compared with BM Mks. We therefore sought to define the immune functions of lung Mks. Using single-cell RNA sequencing of BM and lung myeloid-enriched cells, we found that lung Mks, which we term MkL, had gene expression patterns that are similar to antigen-presenting cells. This was confirmed using imaging and conventional flow cytometry. The immune phenotype of Mks was plastic and driven by the tissue immune environment, as evidenced by BM Mks having an MkL-like phenotype under the influence of pathogen receptor challenge and lung-associated immune molecules, such as IL-33. Our in vitro and in vivo assays demonstrated that MkL internalized and processed both antigenic proteins and bacterial pathogens. Furthermore, MkL induced CD4+ T cell activation in an MHC II-dependent manner both in vitro and in vivo. These data indicated that MkL had key immune regulatory roles dictated in part by the tissue environment.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Pulmão/imunologia , Megacariócitos/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Ativação Linfocitária , Camundongos , Camundongos Knockout , RNA-Seq , Análise de Célula Única
8.
Front Immunol ; 11: 580401, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33381112

RESUMO

Rationale: Protein kinase D (PKD) is a serine/threonine kinase family that is involved in a wide array of signaling pathways. Although PKD has been implicated in immune responses, relatively little is known about the function of PKD in the lung or during viral infections. Objectives: We investigated the hypothesis that PKD is involved in multiple aspects of host response to viral infection. Methods: The selective PKD inhibitor CRT0010166 was administered to C57BL/6 mice prior to and during challenge with either inhaled double-stranded RNA or Influenza A Virus. PKD signaling pathways were investigated in human bronchial epithelial cells treated with CRT0010166, double-stranded RNA, and/or infected with Influenza A Virus. Measurements: Total protein and albumin accumulation in the bronchoalveolar fluid was used to asses inside/out leak. Clearance of inhaled FITC-dextran out of the airspace was used to assess outside/in leak. Cytokines and neutrophils in bronchoalveolar lavage were assayed with ELISAs and cytospins respectively. Viral RNA level was assessed with RT-PCR and protein level assessed by ELISA. Main Results: PKD inhibition prevented airway barrier dysfunction and pro-inflammatory cytokine release. Epithelial cells express PKD3, and PKD3 siRNA knock-down inhibited polyI:C induced cytokine production. Lung epithelial-specific deletion of PKD3 (CC10-Cre x PKD3-floxed mice) partially attenuated polyI:C-induced barrier disruption in vivo. Mechanistically, we found that PKD promoted cytokine mRNA transcription, not secretion, likely through activating the transcription factor Sp1. Finally, prophylactic CRT treatment of mice promoted barrier integrity during influenza virus infection and reduced viral burden. Conclusions: Inhibiting PKD promotes barrier integrity, limit pathogenic cytokine levels, and restrict Influenza A Virus infection. Therefore, PKD is an attractive target for novel antiviral therapeutics.


Assuntos
Vírus da Influenza A/fisiologia , Influenza Humana/imunologia , Infecções por Orthomyxoviridae/imunologia , Proteína Quinase C/metabolismo , Mucosa Respiratória/metabolismo , Animais , Células Cultivadas , Dextranos , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase C/genética , Inibidores de Proteínas Quinases/administração & dosagem , RNA Interferente Pequeno/genética , Mucosa Respiratória/patologia , Transdução de Sinais , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo
9.
PLoS One ; 14(5): e0216056, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31067281

RESUMO

The airway epithelial barrier is critical for preventing pathogen invasion and translocation of inhaled particles into the lung. Epithelial cells also serve an important sentinel role after infection and release various pro-inflammatory mediators that recruit and activate immune cells. Airway epithelial barrier disruption has been implicated in a growing number of respiratory diseases including viral infections. It is thought that when a pathogen breaks the barrier and gains access to the host tissue, pro-inflammatory mediators increase, which further disrupts the barrier and initiates a vicious cycle of leak. However, it is difficult to study airway barrier integrity in vivo, and little is known about relationship between epithelial barrier function and airway inflammation. Current assays of pulmonary barrier integrity quantify the leak of macromolecules from the vasculature into the airspaces (or "inside/out" leak). However, it is also important to measure the ease with which inhaled particles, allergens, or pathogens can enter the subepithelial tissues (or "outside/in" leak). We challenged mice with inhaled double stranded RNA (dsRNA) and explored the relationship between inside/out and outside/in barrier function and airway inflammation. Using wild-type and gene-targeted mice, we studied the roles of the dsRNA sensors Toll Like Receptor 3 (TLR3) and Melanoma Differentiation-Associated protein 5 (MDA5). Here we report that after acute challenge with inhaled dsRNA, airway barrier dysfunction occurs in a TLR3-dependent manner, whereas leukocyte accumulation is largely MDA5-dependent. We conclude that airway barrier dysfunction and inflammation are regulated by different mechanisms at early time points after exposure to inhaled dsRNA.


Assuntos
Inflamação/induzido quimicamente , Helicase IFIH1 Induzida por Interferon/fisiologia , RNA de Cadeia Dupla/farmacologia , Mucosa Respiratória/efeitos dos fármacos , Receptor 3 Toll-Like/fisiologia , Administração por Inalação , Animais , Líquido da Lavagem Broncoalveolar/química , Quimiocina CCL3/análise , Feminino , Inflamação/metabolismo , Inflamação/fisiopatologia , Interferon gama/análise , Interleucina-6/análise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA de Cadeia Dupla/metabolismo , Mucosa Respiratória/metabolismo , Mucosa Respiratória/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa