Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.039
Filtrar
2.
Environ Int ; 166: 107395, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35839670

RESUMO

The anthropogenic particulate matter (PM), suspended air dust that can be inhaled by humans and deposited in the lungs, is one of the main pollutants in the industrialized cities atmosphere. Recent studies have shown that PM has adverse effects on respiratory diseases. These effects are mainly due to the ultrafine particles (PM0.1, PM < 100 nm), which, thanks to their PM size, are efficiently deposited in nasal, tracheobronchial, and alveolar regions. Pulmonary macrophages are a heterogeneous cell population distributed in different lung compartments, whose role in inflammatory response to injury is of particular relevance. In this study, we investigated the effect of PM0.1 on Human Lung Macrophages (HLMs) activation evaluated as proinflammatory cytokines and chemokine release, Reactive Oxygen Species (ROS) production and intracellular Ca2+concentration ([Ca2+]i). Furthermore, PM0.1, after removal of organic fraction, was fractionated in nanoparticles both smaller (NP20) and bigger (NP100) than 20 nm by a properlydeveloped analytical protocol, allowed isolating their individual contribution. Interestingly, while PM0.1 and NP20 induced stimulatory effects on HLM cytokines release, NP100 had not effect. In particular, PM0.1 induced IL-6, IL-1ß, TNF-α, but not CXCL8, release from HLMs. Moreover, PM0.1, NP20 and NP100 did not induce ß-glucuronidase release, a preformed mediator contained in HLMs. The long time necessary for cytokines release (18 h) suggested that PM0.1 and NP20 could induce ex-novo production of the tested mediators. Accordingly, after 6 h of incubation, PM0.1 and NP20 induced mRNA expression of IL-6, TNF-α and IL-1ß. Moreover, NP20 induced ROS production and [Ca2+]i increase in a time-dependent manner, without producing cytotoxicity. Collectively, the present data highlight the main proinflammatory role of NP20 among PM fractions. This is particularly of concern because this fraction is not currently covered by legal limits as it is not easily measured at the exhausts by the available technical methodologies, suggesting that it is mandatory to search for new monitoring techniques and strategies for limiting NP20 formation.


Assuntos
Poluentes Atmosféricos , Macrófagos Alveolares , Material Particulado , Poluentes Atmosféricos/efeitos adversos , Poluentes Atmosféricos/farmacologia , Citocinas/metabolismo , Humanos , Interleucina-6 , Pulmão , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/fisiologia , Tamanho da Partícula , Material Particulado/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
3.
Mediators Inflamm ; 2022: 1002582, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35462787

RESUMO

Background: To examine the role of interferon regulatory factor-1 (IRF-1) and to explore the potential molecular mechanism in ventilator-induced lung injury. Methods: Wild-type C57BL/6 mice and IRF-1 gene knockout mice/caspase-1 knockout mice were mechanically ventilated with a high tidal volume to establish a ventilator-related lung injury model. The supernatant of the alveolar lavage solution and the lung tissues of these mice were collected. The degree of lung injury was examined by hematoxylin and eosin staining. The protein and mRNA expression levels of IRF-1, caspase-1 (p10), and interleukin (IL)-1ß (p17) in lung tissues were measured by western blot and quantitative real-time polymerase chain reaction, respectively. Pyroptosis of alveolar macrophages was detected by flow cytometry and western blotting for active caspase-1 and cleaved GSDMD. An enzyme-linked immunosorbent assay was used to measure the levels of IL-1ß, IL-18, IL-6, TNF-α, and high mobility group box protein 1 (HMGB-1) in alveolar lavage fluid. Results: IRF-1 expression and caspase-1-dependent pyroptosis in lung tissues of wild-type mice were significantly upregulated after mechanical ventilation with a high tidal volume. The degree of ventilator-related lung injury in IRF-1 gene knockout mice and caspase-1 knockout mice was significantly improved compared to that in wild-type mice, and the levels of GSDMD, IL-1ß, IL-18, IL-6, and HMGB-1 in alveolar lavage solution were significantly reduced (P < 0.05). The expression levels of caspase-1 (p10), cleaved GSDMD, and IL-1ß (p17) proteins in lung tissues of IRF-1 knockout mice with ventilator-related lung injury were significantly lower than those of wild-type mice, and the level of pyroptosis of macrophages in alveolar lavage solution was significantly reduced. Conclusions: IRF-1 may aggravate ventilator-induced lung injury by regulating the activation of caspase-1 and the focal death of alveolar macrophages.


Assuntos
Caspase 1 , Fator Regulador 1 de Interferon , Macrófagos Alveolares , Piroptose , Lesão Pulmonar Induzida por Ventilação Mecânica , Animais , Caspase 1/genética , Caspase 1/metabolismo , Proteína HMGB1/metabolismo , Fator Regulador 1 de Interferon/biossíntese , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/metabolismo , Interleucina-18/metabolismo , Interleucina-6/metabolismo , Pulmão/metabolismo , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patologia , Macrófagos Alveolares/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Piroptose/genética , Piroptose/fisiologia , Lesão Pulmonar Induzida por Ventilação Mecânica/genética , Lesão Pulmonar Induzida por Ventilação Mecânica/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/fisiopatologia
4.
Front Immunol ; 13: 773261, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35126390

RESUMO

Short-chain fatty acids (SCFAs) are metabolites produced mainly by the gut microbiota with a known role in immune regulation. Acetate, the major SCFA, is described to disseminate to distal organs such as lungs where it can arm sentinel cells, including alveolar macrophages, to fight against bacterial intruders. In the current study, we explored mechanisms through which acetate boosts macrophages to enhance their bactericidal activity. RNA sequencing analyses show that acetate triggers a transcriptomic program in macrophages evoking changes in metabolic process and immune effector outputs, including nitric oxide (NO) production. In addition, acetate enhances the killing activity of macrophages towards Streptococcus pneumoniae in an NO-dependent manner. Mechanistically, acetate improves IL-1ß production by bacteria-conditioned macrophages and the latter acts in an autocrine manner to promote NO production. Strikingly, acetate-triggered IL-1ß production was neither dependent of its cell surface receptor free-fatty acid receptor 2, nor of the enzymes responsible for its metabolism, namely acetyl-CoA synthetases 1 and 2. We found that IL-1ß production by acetate relies on NLRP3 inflammasome and activation of HIF-1α, the latter being triggered by enhanced glycolysis. In conclusion, we unravel a new mechanism through which acetate reinforces the bactericidal activity of alveolar macrophages.


Assuntos
Citotoxicidade Imunológica , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inflamassomos/metabolismo , Macrófagos Alveolares/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Infecções Pneumocócicas/etiologia , Infecções Pneumocócicas/metabolismo , Streptococcus pneumoniae/imunologia , Acetatos/farmacologia , Animais , Biomarcadores , Citotoxicidade Imunológica/efeitos dos fármacos , Modelos Animais de Doenças , Suscetibilidade a Doenças , Técnicas de Silenciamento de Genes , Glicólise , Interações Hospedeiro-Patógeno/imunologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Interleucina-1beta/metabolismo , Camundongos , Camundongos Knockout , Óxido Nítrico/metabolismo , Consumo de Oxigênio , RNA Interferente Pequeno/genética
5.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35046017

RESUMO

Alveolar macrophages (AMs) are critical for lung immune defense and homeostasis. They are orchestrators of chronic obstructive pulmonary disease (COPD), with their number significantly increased and functions altered in COPD. However, it is unclear how AM number and function are controlled in a healthy lung and if changes in AMs without environmental assault are sufficient to trigger lung inflammation and COPD. We report here that absence of isthmin 1 (ISM1) in mice (Ism1-/- ) leads to increase in both AM number and functional heterogeneity, with enduring lung inflammation, progressive emphysema, and significant lung function decline, phenotypes similar to human COPD. We reveal that ISM1 is a lung resident anti-inflammatory protein that selectively triggers the apoptosis of AMs that harbor high levels of its receptor cell-surface GRP78 (csGRP78). csGRP78 is present at a heterogeneous level in the AMs of a healthy lung, but csGRP78high AMs are expanded in Ism1-/- mice, cigarette smoke (CS)-induced COPD mice, and human COPD lung, making these cells the prime targets of ISM1-mediated apoptosis. We show that csGRP78high AMs mostly express MMP-12, hence proinflammatory. Intratracheal delivery of recombinant ISM1 (rISM1) depleted csGRP78high AMs in both Ism1-/- and CS-induced COPD mice, blocked emphysema development, and preserved lung function. Consistently, ISM1 expression in human lungs positively correlates with AM apoptosis, suggesting similar function of ISM1-csGRP78 in human lungs. Our findings reveal that AM apoptosis regulation is an important physiological mechanism for maintaining lung homeostasis and demonstrate the potential of pulmonary-delivered rISM1 to target csGRP78 as a therapeutic strategy for COPD.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Pulmão/patologia , Macrófagos Alveolares/metabolismo , Células Epiteliais Alveolares/metabolismo , Animais , Apoptose/imunologia , Líquido da Lavagem Broncoalveolar/imunologia , Modelos Animais de Doenças , Chaperona BiP do Retículo Endoplasmático/metabolismo , Chaperona BiP do Retículo Endoplasmático/fisiologia , Feminino , Homeostase , Inflamação , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Pulmão/metabolismo , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fagocitose/fisiologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Enfisema Pulmonar/metabolismo , Fumaça/efeitos adversos , Fumar/efeitos adversos , Nicotiana/efeitos adversos
6.
Front Immunol ; 12: 719727, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34621268

RESUMO

Infectious pneumonia is one of the most common complications after bone marrow transplantation (BMT), which is considered to be associated with poor reconstitution and functional maturation of alveolar macrophages (AMs) post-transplantation. Here, we present evidence showing that lack of IL-13-secreting group 2 innate lymphoid cells (ILC2s) in the lungs may underlay poor AM reconstitution in a mouse model of haploidentical BMT (haplo-BMT). Recombinant murine IL-13 was able to potentiate monocyte-derived AM differentiation in vitro. When intranasally administered, a cocktail of granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-13, and CCL2 not only promoted donor monocyte-derived AM reconstitution in haplo-BMT-recipient mice but also enhanced the innate immunity of the recipient animals against pulmonary bacterial infection. These results provide a useful clue for a clinical strategy to prevent pulmonary bacterial infection at the early stage of recipients post-BMT.


Assuntos
Transplante de Medula Óssea , Diferenciação Celular , Citocinas/metabolismo , Reconstituição Imune , Macrófagos Alveolares/fisiologia , Mielopoese , Animais , Transplante de Medula Óssea/métodos , Diferenciação Celular/efeitos dos fármacos , Citocinas/farmacologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Interleucina-13/metabolismo , Interleucina-13/farmacologia , Linfócitos/citologia , Linfócitos/imunologia , Linfócitos/metabolismo , Macrófagos Alveolares/citologia , Macrófagos Alveolares/efeitos dos fármacos , Camundongos , Modelos Animais , Mielopoese/efeitos dos fármacos , Transplante Haploidêntico
7.
Front Immunol ; 12: 707856, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34335621

RESUMO

Several infectious pathologies in humans, such as tuberculosis or SARS-CoV-2, are responsible for tissue or lung damage, requiring regeneration. The regenerative capacity of adult mammals is limited to few organs. Critical injuries of non-regenerative organs trigger a repair process that leads to a definitive architectural and functional disruption, while superficial wounds result in scar formation. Tissue lesions in mammals, commonly studied under non-infectious conditions, trigger cell death at the site of the injury, as well as the production of danger signals favouring the massive recruitment of immune cells, particularly macrophages. Macrophages are also of paramount importance in infected injuries, characterized by the presence of pathogenic microorganisms, where they must respond to both infection and tissue damage. In this review, we compare the processes implicated in the tissue repair of non-infected versus infected injuries of two organs, the skeletal muscles and the lungs, focusing on the primary role of macrophages. We discuss also the negative impact of infection on the macrophage responses and the possible routes of investigation for new regenerative therapies to improve the recovery state as seen with COVID-19 patients.


Assuntos
COVID-19/imunologia , Macrófagos Alveolares/fisiologia , SARS-CoV-2/fisiologia , Remodelação das Vias Aéreas , Animais , Humanos , Infecções , Mamíferos , Regeneração , Cicatrização
8.
J Exp Med ; 218(10)2021 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-34431978

RESUMO

Programs defining tissue-resident macrophage identity depend on local environmental cues. For alveolar macrophages (AMs), these signals are provided by immune and nonimmune cells and include GM-CSF (CSF2). However, evidence to functionally link components of this intercellular cross talk remains scarce. We thus developed new transgenic mice to profile pulmonary GM-CSF expression, which we detected in both immune cells, including group 2 innate lymphoid cells and γδ T cells, as well as AT2s. AMs were unaffected by constitutive deletion of hematopoietic Csf2 and basophil depletion. Instead, AT2 lineage-specific constitutive and inducible Csf2 deletion revealed the nonredundant function of AT2-derived GM-CSF in instructing AM fate, establishing the postnatal AM compartment, and maintaining AMs in adult lungs. This AT2-AM relationship begins during embryogenesis, where nascent AT2s timely induce GM-CSF expression to support the proliferation and differentiation of fetal monocytes contemporaneously seeding the tissue, and persists into adulthood, when epithelial GM-CSF remains restricted to AT2s.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Pulmão/citologia , Macrófagos Alveolares/fisiologia , Animais , Animais Recém-Nascidos , Diferenciação Celular , Citocinas/metabolismo , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Imunidade Inata , Pulmão/embriologia , Macrófagos Alveolares/citologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
9.
J Nutr Biochem ; 97: 108797, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34126202

RESUMO

Agricultural workers, especially those who work in swine confinement facilities, are at increased risk for developing pulmonary diseases including asthma, chronic obstructive pulmonary disease, and chronic bronchitis due to exposures to fumes, vapors, and organic dust. Repetitive exposure to agricultural dust leads to unresolved inflammation, a common underlying mechanism that worsens lung disease. Besides occupational exposure to dusts, diet also significantly contributes to inflammation and disease progression. Since DHA (docosahexaenoic acid), a polyunsaturated omega-3 fatty acid and its bioactive metabolites have key roles in inflammation resolution, we rationalized that individuals chronically exposed to organic dusts can benefit from dietary modifications. Here, we evaluated the role of DHA in modifying airway inflammation in a murine model of repetitive exposure to an aqueous extract of agricultural dust (three-week exposure to swine confinement dust extract, HDE) and after a one-week resolution/recovery period. We found that mice fed a high DHA diet had significantly increased bronchoalveolar lavage fluid (BALF) levels of DHA-derived resolvins and lower TNFα along with altered plasma levels of endocannabinoids and related lipid mediators. Following the one-week recovery we identified significantly reduced BALF cellularity and cytokine/chemokine release along with increased BALF amphiregulin and resolvins in DHA diet-fed versus control diet-fed mice challenged with HDE. We further report observations on the effects of repetitive HDE exposure on lung Ym1+ and Arg-1+ macrophages. Overall, our findings support a protective role for DHA and identify DHA-derived resolvins and endocannabinoids among the potential mediators of DHA in altering airway inflammation in chronic agricultural dust exposure.


Assuntos
Dieta , Ácidos Docosa-Hexaenoicos/administração & dosagem , Poeira , Exposição por Inalação/efeitos adversos , Doenças Respiratórias/dietoterapia , Doenças dos Trabalhadores Agrícolas/dietoterapia , Doenças dos Trabalhadores Agrícolas/patologia , Animais , Líquido da Lavagem Broncoalveolar/química , Modelos Animais de Doenças , Ácido Eicosapentaenoico/análogos & derivados , Ácido Eicosapentaenoico/metabolismo , Endocanabinoides/sangue , Ácidos Graxos Insaturados/sangue , Inflamação/dietoterapia , Inflamação/patologia , Pulmão/patologia , Macrófagos Alveolares/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doenças Respiratórias/patologia , Suínos , Fator de Necrose Tumoral alfa/metabolismo
10.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-33972447

RESUMO

Pulmonary fibrosis is a relentlessly progressive and often fatal disease with a paucity of available therapies. Genetic evidence implicates disordered epithelial repair, which is normally achieved by the differentiation of small cuboidal alveolar type 2 (AT2) cells into large, flattened alveolar type 1 (AT1) cells as an initiating event in pulmonary fibrosis pathogenesis. Using models of pulmonary fibrosis in young adult and old mice and a model of adult alveologenesis after pneumonectomy, we show that administration of ISRIB, a small molecule that restores protein translation by EIF2B during activation of the integrated stress response (ISR), accelerated the differentiation of AT2 into AT1 cells. Accelerated epithelial repair reduced the recruitment of profibrotic monocyte-derived alveolar macrophages and ameliorated lung fibrosis. These findings suggest a dysfunctional role for the ISR in regeneration of the alveolar epithelium after injury with implications for therapy.


Assuntos
Acetamidas/farmacologia , Células Epiteliais Alveolares/efeitos dos fármacos , Cicloexilaminas/farmacologia , Proteostase/efeitos dos fármacos , Fibrose Pulmonar/tratamento farmacológico , Acetamidas/uso terapêutico , Fatores Etários , Células Epiteliais Alveolares/citologia , Animais , Amianto , Bleomicina , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Cicloexilaminas/uso terapêutico , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteostase/fisiologia , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/patologia , Estresse Fisiológico/efeitos dos fármacos
11.
Ann Clin Lab Sci ; 51(2): 156-162, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33941554

RESUMO

OBJECTIVE: Although coronavirus disease 2019 (COVID-19) typically presents as a respiratory illness, co-existent cardiovascular symptomatology associated with an elevated serum troponin level has been identified as a risk factor for adverse outcomes. Our study addressed the need to correlate serum cardiovascular biomarkers with tissue pathology based on autopsy. MATERIALS AND METHODS: In 13 patients, we reviewed the clinical history and measurements of serum troponin and other biomarkers and correlated them with autopsy findings. RESULTS: At autopsy, the 13 COVID-19 patients exhibited evidence of diffuse alveolar damage (DAD) and cardiomegaly (heart weights ranged from 380 to 1170 grams). Of the 13 patients, three had elevated troponin I and evidence of severe coronary artery disease (CAD) (cases 4, 5, and 11), while six had elevated troponin I without evidence of severe CAD (cases 1, 3, 6, 7, 8, and 9), and four had no clinical or pathological evidence of CAD. Of note, cases 7 and 9 had significantly elevated troponin I levels (8.84 ng/mL and 4.94 ng/mL, respectively). Several cases showed focal degenerative change or damage of cardiomyocytes. However, none of the cases had evidence of lymphocytic myocarditis. CONCLUSION: Although we observed elevated biomarkers of heart failure in some cases, it was not a consistent finding and did not correlate with evidence of myocarditis. The elevated biomarkers may reflect non-ischemic heart damage as a consequence of COVID-19 infection.


Assuntos
Biomarcadores/sangue , COVID-19 , Cardiomegalia , Pulmão , Macrófagos Alveolares , Miocárdio/patologia , SARS-CoV-2/isolamento & purificação , Autopsia/métodos , Autopsia/estatística & dados numéricos , COVID-19/mortalidade , COVID-19/patologia , Cardiomegalia/patologia , Cardiomegalia/virologia , Movimento Celular , Feminino , Insuficiência Cardíaca/sangue , Humanos , Pulmão/patologia , Pulmão/virologia , Macrófagos Alveolares/patologia , Macrófagos Alveolares/fisiologia , Macrófagos Alveolares/virologia , Masculino , Pessoa de Meia-Idade , Peptídeo Natriurético Encefálico/sangue , Tamanho do Órgão , Fragmentos de Peptídeos/sangue , Troponina I/sangue
12.
Int J Radiat Oncol Biol Phys ; 110(2): 526-538, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33385497

RESUMO

PURPOSE: Type II pneumocyte (alveolar epithelial cells type II [AECII]) senescence has been implicated in the progression of lung fibrosis. The capacity of senescent cells to modulate pulmonary macrophages to drive fibrosis is unexplored. Insulin-like growth factor-1 receptor (IGF-1R) signaling has been implicated as a regulator of senescence and aging. METHODS AND MATERIALS: Mice with an AECII-specific deletion of IGF-1R received thoracic irradiation (n ≥ 5 per condition), and the effect of IGF-1R deficiency on radiation-induced AECII senescence and macrophage polarization to an alternatively activated phenotype (M2) was investigated. IGF-1R signaling, macrophage polarization, and senescence were evaluated in surgically resected human lung (n = 63). RESULTS: IGF-1R deficient mice demonstrated reduced AECII senescence (senescent AECII/field; intact: 7.25% ± 3.5% [mean ± SD], deficient: 2.75% ± 2.8%, P = .0001), reduced accumulation of M2 macrophages (intact: 24.7 ± 2.2 cells/field, deficient: 15.5 ± 1.2 cells/field, P = .0086), and fibrosis (hydroxyproline content; intact: 71.9 ± 21.7 µg/lung, deficient: 31.7 ± 7.9, P = .0485) after irradiation. Senescent AECII enhanced M2 polarization in a paracrine fashion (relative Arg1 mRNA, 0 Gy: 1.0 ± 0.4, 17.5 Gy: 7.34 ± 0.5, P < .0001). Evaluation of surgical samples from patients treated with chemoradiation demonstrated increased expression of IGF-1 (unirradiated: 10.2% ± 4.9% area, irradiated: 15.1% ± 11.5%, P = .0377), p21 (unirradiated: 0.013 ± 0.02 histoscore, irradiated: 0.084 ± 0.09 histoscore, P = .0002), IL-13 (unirradiated: 13.7% ± 2.8% area, irradiated: 21.7% ± 3.8%, P < .0001), and M2 macrophages in fibrotic regions relative to nonfibrotic regions (unirradiated: 11.4 ± 12.2 CD163 + cells/core, irradiated: 43.1 ± 40.9 cells/core, P = .0011), consistent with findings from animal models of lung fibrosis. CONCLUSIONS: This study demonstrates that senescent AECII are necessary for the progression of pulmonary fibrosis and serve as a targetable, chronic stimuli for macrophage activation in fibrotic lung.


Assuntos
Células Epiteliais Alveolares/fisiologia , Polaridade Celular , Senescência Celular/fisiologia , Macrófagos Alveolares/fisiologia , Fibrose Pulmonar/etiologia , Receptor IGF Tipo 1/metabolismo , Células Epiteliais Alveolares/efeitos da radiação , Animais , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/terapia , Senescência Celular/efeitos da radiação , Quimiorradioterapia , Deleção de Genes , Humanos , Hidroxiprolina/análise , Pulmão/metabolismo , Pulmão/efeitos da radiação , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Ativação de Macrófagos , Macrófagos Alveolares/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Fibrose Pulmonar/patologia , Lesões Experimentais por Radiação/metabolismo , Lesões Experimentais por Radiação/fisiopatologia , Lesões Experimentais por Radiação/prevenção & controle , Receptor IGF Tipo 1/deficiência , Receptor IGF Tipo 1/genética
13.
J Allergy Clin Immunol ; 147(4): 1242-1260, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32910932

RESUMO

BACKGROUND: Fine-tuning of immune receptor signaling is critical for the development and functioning of immune cells. Moreover, GM-CSF receptor (GM-CSFR) signaling plays an essential role in the development of certain myeloid lineage cells, including alveolar macrophages (AMs). However, the significance of fine-tuning of GM-CSFR signaling in AMs and its relevance in allergic inflammation have not been reported. OBJECTIVE: Our aim was to explore whether phosphatase Ssu72, originally identified as a regulator of RNA polymerase II activity, regulates AM development and allergic airway inflammation by regulating GM-CSF signaling. METHODS: To address these issues, we generated LysM-CreSsu72fl/fl and Cd11c-CreSsu72fl/fl mice and used ovalbumin- or house dust mite-induced allergic asthma models. RESULTS: Following GM-CSF stimulation, Ssu72 directly bound to the GM-CSFR ß-chain in AMs, preventing phosphorylation. Consistently, mature Ssu72-deficient AMs showed higher phosphorylation of the GM-CSFR ß-chain and downstream molecules, which resulted in greater dysregulation of cell cycle, cell death, cell turnover, mitochondria-related metabolism, and LPS responsiveness in AMs than in mature wild-type AMs. The dysregulation was restored by using a Janus kinase 2 inhibitor, which reduced GM-CSFR ß-chain phosphorylation. LysM-CreSsu72fl/fl mice exhibited deficits in development and maturation of AMs, which were also seen postnatally in Cd11c-CreSsu72fl/fl mice. Furthermore, LysM-CreSsu72fl/fl mice were less responsive to ovalbumin- or house dust mite-induced allergic asthma models than the control mice were; however, their responsiveness was restored by adoptive transfer of JAK2 inhibitor-pretreated mature Ssu72-deficient AMs. CONCLUSION: Our results demonstrate that Ssu72 fine-tunes GM-CSFR signaling by both binding to and reducing phosphorylation of GM-CSFR ß-chain, thereby regulating the development, maturation, and mitochondrial functions of AMs and allergic airway inflammation.


Assuntos
Hipersensibilidade/imunologia , Macrófagos Alveolares/fisiologia , Fosfoproteínas Fosfatases/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Hipersensibilidade Respiratória/imunologia , Animais , Antígenos de Dermatophagoides/imunologia , Antígeno CD11c/metabolismo , Diferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovalbumina/imunologia , Fosfoproteínas Fosfatases/genética , Pyroglyphidae , Transdução de Sinais
14.
Life Sci ; 264: 118617, 2021 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-33096114

RESUMO

BACKGROUND: COVID-19-associated acute respiratory distress syndrome (ARDS) is associated with significant morbidity and high levels of mortality. This paper describes the processes involved in the pathophysiology of COVID-19 from the initial infection and subsequent destruction of type II alveolar epithelial cells by SARS-CoV-2 and culminating in the development of ARDS. MAIN BODY: The activation of alveolar cells and alveolar macrophages leads to the release of large quantities of proinflammatory cytokines and chemokines and their translocation into the pulmonary vasculature. The presence of these inflammatory mediators in the vascular compartment leads to the activation of vascular endothelial cells platelets and neutrophils and the subsequent formation of platelet neutrophil complexes. These complexes in concert with activated endothelial cells interact to create a state of immunothrombosis. The consequence of immunothrombosis include hypercoagulation, accelerating inflammation, fibrin deposition, migration of neutrophil extracellular traps (NETs) producing neutrophils into the alveolar apace, activation of the NLRP3 inflammazome, increased alveolar macrophage destruction and massive tissue damage by pyroptosis and necroptosis Therapeutic combinations aimed at ameliorating immunothrombosis and preventing the development of severe COVID-19 are discussed in detail.


Assuntos
COVID-19/imunologia , COVID-19/fisiopatologia , Síndrome do Desconforto Respiratório/complicações , Síndrome do Desconforto Respiratório/prevenção & controle , SARS-CoV-2/patogenicidade , Trombose/complicações , Trombose/fisiopatologia , Células Epiteliais Alveolares/fisiologia , Plaquetas/fisiologia , COVID-19/complicações , Citocinas/fisiologia , Células Endoteliais/fisiologia , Humanos , Macrófagos Alveolares/fisiologia , Neutrófilos/fisiologia , Síndrome do Desconforto Respiratório/imunologia , Síndrome do Desconforto Respiratório/patologia , Trombose/imunologia , Tratamento Farmacológico da COVID-19
15.
Am J Respir Crit Care Med ; 203(10): 1230-1244, 2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-33306940

RESUMO

Rationale: Tissue-resident memory T cells (TRM) play a critical role in the defense against inhaled pathogens. The isolation and study of human lung tissue-resident memory T cells and lung-resident macrophages (MLR) are limited by experimental constraints. Objectives: To characterize the spatial and functional relationship between MLR and human lung tissue-resident memory T cells using ex vivo lung perfusion (EVLP). Methods: TRM and MLR were isolated using EVLP and intraperfusate-labeled CD45 antibody. Cells isolated after 6 hours of EVLP were analyzed using spectral flow cytometry. Spatial relationships between CD3+ and CD68+ cells were explored with multiplexed immunohistochemistry. Functional relationships were determined by using coculture and T-cell-receptor complex signal transduction. Measurements and Main Results: Lungs from 8 research-consenting organ donors underwent EVLP for 6 hours. We show that human lung TRM and MLR colocalize within the human lung, preferentially around the airways. Furthermore, we found that human lung CD8+ TRM are composed of two functionally distinct populations on the basis of PD1 (programed cell death receptor 1) and ZNF683 (HOBIT) protein expression. We show that MLR provide costimulatory signaling to PD1hi CD4+ and CD8+ lung TRM,, augmenting the effector cytokine production and degranulation of TRM. Conclusions: EVLP provides an innovative technique to study resident immune populations in humans. Human MLR colocalize with and provide costimulation signaling to TRM, augmenting their effector function.


Assuntos
Linfócitos T CD4-Positivos/fisiologia , Linfócitos T CD8-Positivos/fisiologia , Memória Imunológica/fisiologia , Pulmão/imunologia , Pulmão/patologia , Macrófagos Alveolares/fisiologia , Adulto , Técnicas de Cocultura , Citocinas/metabolismo , Feminino , Humanos , Transplante de Pulmão , Masculino , Pessoa de Meia-Idade , Perfusão , Técnicas de Cultura de Tecidos
16.
Cell Rep ; 33(13): 108553, 2020 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-33378665

RESUMO

There is an increasing appreciation for the heterogeneity of myeloid lineages in the lung, but relatively little is known about populations specifically associated with the conducting airways. We use single-cell RNA sequencing, flow cytometry, and immunofluorescence to characterize myeloid cells of the mouse trachea during homeostasis and epithelial injury/repair. We identify submucosal macrophages, similar to lung interstitial macrophages, and intraepithelial macrophages. Following injury, there are early increases in neutrophils and submucosal macrophages, including M2-like macrophages. Intraepithelial macrophages are lost after injury and later restored by CCR2+ monocytes. We show that repair of the tracheal epithelium is impaired in Ccr2-deficient mice. Mast cells and group 2 innate lymphoid cells are sources of interleukin-13 (IL-13) that polarize macrophages and directly influence basal cell behaviors. Their proximity to the airway epithelium establishes these myeloid populations as potential therapeutic targets for airway disease.


Assuntos
Células Epiteliais/metabolismo , Epitélio/metabolismo , Homeostase , Macrófagos Alveolares/fisiologia , Células Mieloides/fisiologia , Receptores CCR2/metabolismo , Traqueia/metabolismo , Animais , Células Cultivadas , Citocinas/metabolismo , Epitélio/lesões , Feminino , Pulmão/metabolismo , Lesão Pulmonar/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Monócitos/metabolismo , Polidocanol , Receptores CCR2/genética , Regeneração , Análise de Sequência de RNA , Análise de Célula Única , Traqueia/lesões
17.
JCI Insight ; 5(24)2020 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-33141765

RESUMO

Alveolar macrophages (AMs) are differentially regulated by human surfactant protein-A1 (SP-A1) or SP-A2. However, AMs are very heterogeneous and differences are difficult to characterize in intact cells. Using the Toponome Imaging System (TIS), an imaging technique that uses sequential immunostaining to identify patterns of biomarker expression or combinatorial molecular phenotypes (CMPs), we studied individual single cells and identified subgroups of AMs (n = 168) from SP-A-KO mice and mice expressing either SP-A1 or SP-A2. The effects, as shown by CMPs, of SP-A1 and SP-A2 on AMs were significant and differed. SP-A1 AMs were the most diverse and shared the fewest CMPs with KO and SP-A2. Clustering analysis of each group showed 3 clusters where the CMP-based phenotype was distinct in each cluster. Moreover, a clustering analysis of all 168 AMs revealed 10 clusters, many dominated by 1 group. Some CMP overlap among groups was observed with SP-A2 AMs sharing the most CMPs and SP-A1 AMs the fewest. The CMP-based patterns identified here provide a basis for understanding not only AMs' diversity, but also most importantly, the molecular basis for the diversity of functional differences in mouse models where the impact of genetics of innate immune molecules on AMs has been studied.


Assuntos
Macrófagos Alveolares/metabolismo , Proteína A Associada a Surfactante Pulmonar/metabolismo , Animais , Humanos , Imunidade Inata/fisiologia , Macrófagos Alveolares/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência/métodos , Proteoma/metabolismo , Proteína A Associada a Surfactante Pulmonar/genética , Proteína A Associada a Surfactante Pulmonar/fisiologia , Surfactantes Pulmonares/metabolismo
18.
Vet Microbiol ; 250: 108851, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33002681

RESUMO

Infection by enveloped viruses includes endocytosis and/or membrane fusion at the plasma membrane, where host cell proteases play an essential role. Among them, elastase-mediated infection has been documented for several enveloped viruses. Porcine reproductive and respiratory syndrome virus (PRRSV), an economically critical factor in global swine industry, is previously reported to infect host cells via low pH-dependent clathrin-mediated endocytosis (CME) and undergo membrane fusion in recycling endosomes. In the current study, we identified that elastase was significantly elevated in the lung tissues of highly pathogenic PRRSV (HP-PRRSV)-infected pigs compared to the mock-infected ones. We subsequently demonstrated that elastase contributed to HP-PRRSV infection in both MARC-145 cells and porcine alveolar macrophages (PAMs). Mechanistically, HP-PRRSV entered host cells at the cell surface via elastase-mediated membrane fusion, independent of low pH and CME, and its glycoprotein 5 (GP5) was cleaved by the protease during this process. All these findings deepen our understanding of HP-PRRSV infection, and are beneficial for prevention and control of the disease.


Assuntos
Interações entre Hospedeiro e Microrganismos , Macrófagos Alveolares/virologia , Fusão de Membrana , Elastase Pancreática/genética , Vírus da Síndrome Respiratória e Reprodutiva Suína/fisiologia , Vírus da Síndrome Respiratória e Reprodutiva Suína/patogenicidade , Animais , Linhagem Celular , Células HEK293 , Humanos , Pulmão/virologia , Macrófagos Alveolares/fisiologia , Elastase Pancreática/metabolismo , Organismos Livres de Patógenos Específicos , Suínos , Doenças dos Suínos/virologia , Internalização do Vírus
19.
J Immunol Res ; 2020: 5097920, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33123603

RESUMO

In this study, the effects of single immunoglobin IL-1 receptor-related protein (SIGIRR) on tumor necrosis factor- (TNF-) receptor-associated factor 6 (TRAF6) ubiquitination in acute lung injury (ALI) were evaluated in both alveolar epithelial cells and alveolar macrophage cells in vitro. Our results found that SIGIRR negatively regulated TRAF6 ubiquitination and such SIGIRR inhibition could enhance the TRAF6 expression in both alveolar epithelial cells (AECs) and alveolar macrophage cells (AMCs). SIGIRR knockdown may increase NF-κB activity via TRAF6 regulation by the classical but not the nonclassical NF-κB signaling pathway. Such modulation between TRAF6 and SIGIRR could affect cytokine secretion and exacerbate the immune response; the IL-8, NFKB1, and NFKBIA mRNA levels were reduced after SIGIRR overexpression. The current study reveals the molecular mechanisms of the negative regulatory roles of SIGIRR on the innate immune response related to the LPS/TLR-4 signaling pathway and provides evidence for strategies to clinically treat inflammatory diseases.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Células Epiteliais Alveolares/fisiologia , Macrófagos Alveolares/fisiologia , Receptores de Interleucina-1/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Células Cultivadas , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Imunidade Inata , Imunomodulação , Interleucina-8/metabolismo , Lipopolissacarídeos/imunologia , NF-kappa B/metabolismo , Receptores de Interleucina-1/genética , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo , Ubiquitinação
20.
J Immunol ; 205(6): 1601-1607, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32796026

RESUMO

Secondary Streptococcus pneumoniae infection is a significant cause of morbidity and mortality during influenza epidemics and pandemics. Multiple pathogenic mechanisms, such as lung epithelial damage and dysregulation of neutrophils and alveolar macrophages (AMs), have been suggested to contribute to the severity of disease. However, the fundamental reasons for influenza-induced susceptibility to secondary bacterial pneumonia remain unclear. In this study, we revisited these controversies over key pathogenic mechanisms in a lethal model of secondary bacterial pneumonia with an S. pneumoniae strain that is innocuous to mice in the absence of influenza infection. Using a series of in vivo models, we demonstrate that rather than a systemic suppression of immune responses or neutrophil function, influenza infection activates IFN-γR signaling and abrogates AM-dependent bacteria clearance and thereby causes extreme susceptibility to pneumococcal infection. Importantly, using mice carrying conditional knockout of Ifngr1 gene in different myeloid cell subsets, we demonstrate that influenza-induced IFN-γR signaling in AMs impairs their antibacterial function, thereby enabling otherwise noninvasive S. pneumoniae to cause deadly pneumonia.


Assuntos
Vírus da Influenza A/fisiologia , Influenza Humana/imunologia , Macrófagos Alveolares/fisiologia , Infecções por Orthomyxoviridae/imunologia , Pneumonia Pneumocócica/imunologia , Receptores de Interferon/metabolismo , Streptococcus pneumoniae/fisiologia , Animais , Coinfecção , Modelos Animais de Doenças , Suscetibilidade a Doenças , Humanos , Tolerância Imunológica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Interferon/genética , Transdução de Sinais , Receptor de Interferon gama
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...