Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 185(5): 815-830.e19, 2022 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-35148838

RESUMO

Rapid neutrophil recruitment to sites of inflammation is crucial for innate immune responses. Here, we reveal that the G-protein-coupled receptor GPR35 is upregulated in activated neutrophils, and it promotes their migration. GPR35-deficient neutrophils are less recruited from blood vessels into inflamed tissue, and the mice are less efficient in clearing peritoneal bacteria. Using a bioassay, we find that serum and activated platelet supernatant stimulate GPR35, and we identify the platelet-derived serotonin metabolite 5-hydroxyindoleacetic acid (5-HIAA) as a GPR35 ligand. GPR35 function in neutrophil recruitment is strongly dependent on platelets, with the receptor promoting transmigration across platelet-coated endothelium. Mast cells also attract GPR35+ cells via 5-HIAA. Mice deficient in 5-HIAA show a loss of GPR35-mediated neutrophil recruitment to inflamed tissue. These findings identify 5-HIAA as a GPR35 ligand and neutrophil chemoattractant and establish a role for platelet- and mast cell-produced 5-HIAA in cell recruitment to the sites of inflammation and bacterial clearance.


Assuntos
Ácido Hidroxi-Indolacético/metabolismo , Neutrófilos , Receptores Acoplados a Proteínas G/metabolismo , Animais , Inflamação/metabolismo , Ligantes , Camundongos , Infiltração de Neutrófilos , Neutrófilos/metabolismo , Serotonina/metabolismo
3.
Nature ; 591(7848): 124-130, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33494096

RESUMO

Although infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has pleiotropic and systemic effects in some individuals1-3, many others experience milder symptoms. Here, to gain a more comprehensive understanding of the distinction between severe and mild phenotypes in the pathology of coronavirus disease 2019 (COVID-19) and its origins, we performed a whole-blood-preserving single-cell analysis protocol to integrate contributions from all major immune cell types of the blood-including neutrophils, monocytes, platelets, lymphocytes and the contents of the serum. Patients with mild COVID-19 exhibit a coordinated pattern of expression of interferon-stimulated genes (ISGs)3 across every cell population, whereas these ISG-expressing cells are systemically absent in patients with severe disease. Paradoxically, individuals with severe COVID-19 produce very high titres of anti-SARS-CoV-2 antibodies and have a lower viral load compared to individuals with mild disease. Examination of the serum from patients with severe COVID-19 shows that these patients uniquely produce antibodies that functionally block the production of the ISG-expressing cells associated with mild disease, by activating conserved signalling circuits that dampen cellular responses to interferons. Overzealous antibody responses pit the immune system against itself in many patients with COVID-19, and perhaps also in individuals with other viral infections. Our findings reveal potential targets for immunotherapies in patients with severe COVID-19 to re-engage viral defence.


Assuntos
Anticorpos Antivirais/imunologia , COVID-19/imunologia , COVID-19/fisiopatologia , Interferons/antagonistas & inibidores , Interferons/imunologia , SARS-CoV-2/imunologia , SARS-CoV-2/patogenicidade , Anticorpos Antivirais/sangue , Formação de Anticorpos , Sequência de Bases , COVID-19/sangue , COVID-19/virologia , Feminino , Humanos , Imunoglobulina G/imunologia , Interferons/metabolismo , Masculino , Neutrófilos/imunologia , Neutrófilos/patologia , Domínios Proteicos , Receptor de Interferon alfa e beta/antagonistas & inibidores , Receptor de Interferon alfa e beta/imunologia , Receptor de Interferon alfa e beta/metabolismo , Receptores de IgG/imunologia , Análise de Célula Única , Carga Viral/imunologia
4.
Am J Respir Crit Care Med ; 209(1): 70-82, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37878820

RESUMO

Rationale: Acute lung injury (ALI) carries a high risk of mortality but has no established pharmacologic therapy. We previously found that experimental ALI occurs through natural killer (NK) cell NKG2D receptor activation and that the cognate human ligand, MICB, was associated with ALI after transplantation. Objectives: To investigate the association of a common missense variant, MICBG406A, with ALI. Methods: We assessed MICBG406A genotypes within two multicenter observational study cohorts at risk for ALI: primary graft dysfunction (N = 619) and acute respiratory distress syndrome (N = 1,376). Variant protein functional effects were determined in cultured and ex vivo human samples. Measurements and Main Results: Recipients of MICBG406A-homozygous allografts had an 11.1% absolute risk reduction (95% confidence interval [CI], 3.2-19.4%) for severe primary graft dysfunction after lung transplantation and reduced risk for allograft failure (hazard ratio, 0.36; 95% CI, 0.13-0.98). In participants with sepsis, we observed 39% reduced odds of moderately or severely impaired oxygenation among MICBG406A-homozygous individuals (95% CI, 0.43-0.86). BAL NK cells were less frequent and less mature in participants with MICBG406A. Expression of missense variant protein MICBD136N in cultured cells resulted in reduced surface MICB and reduced NKG2D ligation relative to wild-type MICB. Coculture of variant MICBD136N cells with NK cells resulted in less NKG2D activation and less susceptibility to NK cell killing relative to the wild-type cells. Conclusions: These data support a role for MICB signaling through the NKG2D receptor in mediating ALI, suggesting a novel therapeutic approach.


Assuntos
Lesão Pulmonar Aguda , Disfunção Primária do Enxerto , Humanos , Lesão Pulmonar Aguda/genética , Genômica , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo
5.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-34244428

RESUMO

The emerging field of regenerative cell therapy is still limited by the few cell types that can reliably be differentiated from pluripotent stem cells and by the immune hurdle of commercially scalable allogeneic cell therapeutics. Here, we show that gene-edited, immune-evasive cell grafts can survive and successfully treat diseases in immunocompetent, fully allogeneic recipients. Transplanted endothelial cells improved perfusion and increased the likelihood of limb preservation in mice with critical limb ischemia. Endothelial cell grafts transduced to express a transgene for alpha1-antitrypsin (A1AT) successfully restored physiologic A1AT serum levels in mice with genetic A1AT deficiency. This cell therapy prevented both structural and functional changes of emphysematous lung disease. A mixture of endothelial cells and cardiomyocytes was injected into infarcted mouse hearts, and both cell types orthotopically engrafted in the ischemic areas. Cell therapy led to an improvement in invasive hemodynamic heart failure parameters. Our study supports the development of hypoimmune, universal regenerative cell therapeutics for cost-effective treatments of major diseases.


Assuntos
Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/terapia , Imunocompetência , Células-Tronco Pluripotentes Induzidas/imunologia , Pneumopatias/imunologia , Pneumopatias/terapia , Transplante de Células-Tronco , Animais , Células Endoteliais/transplante , Insuficiência Cardíaca/terapia , Membro Posterior/irrigação sanguínea , Membro Posterior/patologia , Isquemia/patologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/transplante , Transplante Homólogo , alfa 1-Antitripsina/metabolismo
6.
Am J Physiol Lung Cell Mol Physiol ; 325(2): L262-L269, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37401383

RESUMO

Microbes, toxins, therapeutics, and cells are often instilled into lungs of mice to model diseases and test experimental interventions. Consistent pulmonary delivery is critical for experimental power and reproducibility, but we observed variation in outcomes between handlers using different anesthetic approaches for intranasal dosing in mice. We therefore used a radiotracer to quantify lung delivery after intranasal dosing under inhalational (isoflurane) versus injectable (ketamine/xylazine) anesthesia in C57BL/6 mice. We found that ketamine/xylazine anesthesia resulted in delivery of a greater proportion (52 ± 9%) of an intranasal dose to lungs relative to isoflurane anesthesia (30 ± 15%). This difference in pulmonary dose delivery altered key outcomes in models of viral and bacterial pneumonia, with mice anesthetized with ketamine/xylazine for intranasal infection with influenza A virus or Pseudomonas aeruginosa developing more robust lung inflammation responses relative to control animals randomized to isoflurane anesthesia. Pulmonary dosing efficiency through oropharyngeal aspiration was not affected by anesthetic method and resulted in delivery of 63 ± 8% of dose to lungs, and a nonsurgical intratracheal dosing approach further increased lung delivery to 92 ± 6% of dose. The use of either of these more precise dosing methods yielded greater experimental power in the bacterial pneumonia model relative to intranasal infection. Both anesthetic approach and dosing route can impact pulmonary dosing efficiency. These factors affect experimental power and so should be considered when planning and reporting studies involving delivery of fluids to lungs of mice.NEW & NOTEWORTHY Many lung research studies involve dosing fluids into lungs of mice. In this study, the authors measure lung deposition using intranasal (i.n.), oropharyngeal aspiration (o.a.), and intratracheal (i.t.) dosing methods in mice. Anesthetic approach and administration route were found to affect pulmonary dosing efficiency. The authors demonstrate that refinements to dosing techniques can enable reductions in the number of animals needed for bacterial and viral pneumonia studies.


Assuntos
Anestesia , Anestésicos , Isoflurano , Ketamina , Animais , Camundongos , Anestesia/métodos , Pulmão , Camundongos Endogâmicos C57BL , Reprodutibilidade dos Testes , Xilazina
7.
Circ Res ; 128(5): 655-669, 2021 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-33508948

RESUMO

RATIONALE: Circulating monocytes can have proinflammatory or proreparative phenotypes. The endogenous signaling molecules and pathways that regulate monocyte polarization in vivo are poorly understood. We have shown that platelet-derived ß2M (ß-2 microglobulin) and TGF-ß (transforming growth factor ß) have opposing effects on monocytes by inducing inflammatory and reparative phenotypes, respectively, but each bind and signal through the same receptor. We now define the signaling pathways involved. OBJECTIVE: To determine the molecular mechanisms and signal transduction pathways by which ß2M and TGF-ß regulate monocyte responses both in vitro and in vivo. METHODS AND RESULTS: Wild-type- (WT) and platelet-specific ß2M knockout mice were treated intravenously with either ß2M or TGF-ß to increase plasma concentrations to those in cardiovascular diseases. Elevated plasma ß2M increased proinflammatory monocytes, while increased plasma TGFß increased proreparative monocytes. TGF-ßR (TGF-ß receptor) inhibition blunted monocyte responses to both ß2M and TGF-ß in vivo. Using imaging flow cytometry, we found that ß2M decreased monocyte SMAD2/3 nuclear localization, while TGF-ß promoted SMAD nuclear translocation but decreased noncanonical/inflammatory (JNK [jun kinase] and NF-κB [nuclear factor-κB] nuclear localization). This was confirmed in vitro using both imaging flow cytometry and immunoblots. ß2M, but not TGF-ß, promoted ubiquitination of SMAD3 and SMAD4, that inhibited their nuclear trafficking. Inhibition of ubiquitin ligase activity blocked noncanonical SMAD-independent monocyte signaling and skewed monocytes towards a proreparative monocyte response. CONCLUSIONS: Our findings indicate that elevated plasma ß2M and TGF-ß dichotomously polarize monocytes. Furthermore, these immune molecules share a common receptor but induce SMAD-dependent canonical signaling (TGF-ß) versus noncanonical SMAD-independent signaling (ß2M) in a ubiquitin ligase dependent manner. This work has broad implications as ß2M is increased in several inflammatory conditions, while TGF-ß is increased in fibrotic diseases. Graphic Abstract: A graphic abstract is available for this article.


Assuntos
Monócitos/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Microglobulina beta-2/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Humanos , MAP Quinase Quinase 4/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/citologia , Monócitos/efeitos dos fármacos , NF-kappa B/metabolismo , Proteínas Smad/metabolismo , Células THP-1 , Microglobulina beta-2/farmacologia
8.
Nature ; 544(7648): 105-109, 2017 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-28329764

RESUMO

Platelets are critical for haemostasis, thrombosis, and inflammatory responses, but the events that lead to mature platelet production remain incompletely understood. The bone marrow has been proposed to be a major site of platelet production, although there is indirect evidence that the lungs might also contribute to platelet biogenesis. Here, by directly imaging the lung microcirculation in mice, we show that a large number of megakaryocytes circulate through the lungs, where they dynamically release platelets. Megakaryocytes that release platelets in the lungs originate from extrapulmonary sites such as the bone marrow; we observed large megakaryocytes migrating out of the bone marrow space. The contribution of the lungs to platelet biogenesis is substantial, accounting for approximately 50% of total platelet production or 10 million platelets per hour. Furthermore, we identified populations of mature and immature megakaryocytes along with haematopoietic progenitors in the extravascular spaces of the lungs. Under conditions of thrombocytopenia and relative stem cell deficiency in the bone marrow, these progenitors can migrate out of the lungs, repopulate the bone marrow, completely reconstitute blood platelet counts, and contribute to multiple haematopoietic lineages. These results identify the lungs as a primary site of terminal platelet production and an organ with considerable haematopoietic potential.


Assuntos
Plaquetas/citologia , Hematopoese , Células-Tronco Hematopoéticas/citologia , Pulmão/irrigação sanguínea , Pulmão/citologia , Animais , Medula Óssea , Linhagem da Célula , Feminino , Pulmão/anatomia & histologia , Masculino , Megacariócitos/citologia , Camundongos , Microcirculação , Contagem de Plaquetas , Trombocitopenia/patologia
9.
Am J Respir Cell Mol Biol ; 67(3): 284-308, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35679511

RESUMO

Clinical and molecular heterogeneity are common features of human disease. Understanding the basis for heterogeneity has led to major advances in therapy for many cancers and pulmonary diseases such as cystic fibrosis and asthma. Although heterogeneity of risk factors, disease severity, and outcomes in survivors are common features of the acute respiratory distress syndrome (ARDS), many challenges exist in understanding the clinical and molecular basis for disease heterogeneity and using heterogeneity to tailor therapy for individual patients. This report summarizes the proceedings of the 2021 Aspen Lung Conference, which was organized to review key issues related to understanding clinical and molecular heterogeneity in ARDS. The goals were to review new information about ARDS phenotypes, to explore multicellular and multisystem mechanisms responsible for heterogeneity, and to review how best to account for clinical and molecular heterogeneity in clinical trial design and assessment of outcomes. The report concludes with recommendations for future research to understand the clinical and basic mechanisms underlying heterogeneity in ARDS to advance the development of new treatments for this life-threatening critical illness.


Assuntos
Síndrome do Desconforto Respiratório , Humanos , Pulmão , Fatores de Risco , Índice de Gravidade de Doença , Tórax
10.
Am J Respir Cell Mol Biol ; 66(2): e1-e14, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35103557

RESUMO

Advancements in methods, technology, and our understanding of the pathobiology of lung injury have created the need to update the definition of experimental acute lung injury (ALI). We queried 50 participants with expertise in ALI and acute respiratory distress syndrome using a Delphi method composed of a series of electronic surveys and a virtual workshop. We propose that ALI presents as a "multidimensional entity" characterized by four "domains" that reflect the key pathophysiologic features and underlying biology of human acute respiratory distress syndrome. These domains are 1) histological evidence of tissue injury, 2) alteration of the alveolar-capillary barrier, 3) presence of an inflammatory response, and 4) physiologic dysfunction. For each domain, we present "relevant measurements," defined as those proposed by at least 30% of respondents. We propose that experimental ALI encompasses a continuum of models ranging from those focusing on gaining specific mechanistic insights to those primarily concerned with preclinical testing of novel therapeutics or interventions. We suggest that mechanistic studies may justifiably focus on a single domain of lung injury, but models must document alterations of at least three of the four domains to qualify as "experimental ALI." Finally, we propose that a time criterion defining "acute" in ALI remains relevant, but the actual time may vary based on the specific model and the aspect of injury being modeled. The continuum concept of ALI increases the flexibility and applicability of the definition to multiple models while increasing the likelihood of translating preclinical findings to critically ill patients.


Assuntos
Lesão Pulmonar Aguda/patologia , Inflamação/fisiopatologia , Relatório de Pesquisa/tendências , Lesão Pulmonar Aguda/imunologia , Animais
11.
Nature ; 531(7595): 513-7, 2016 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-26982733

RESUMO

Lung metastasis is the lethal determinant in many cancers and a number of lines of evidence point to monocytes and macrophages having key roles in its development. Yet little is known about the immediate fate of incoming tumour cells as they colonize this tissue, and even less known about how they make first contact with the immune system. Primary tumours liberate circulating tumour cells (CTCs) into the blood and we have developed a stable intravital two-photon lung imaging model in mice for direct observation of the arrival of CTCs and subsequent host interaction. Here we show dynamic generation of tumour microparticles in shear flow in the capillaries within minutes of CTC entry. Rather than dispersing under flow, many of these microparticles remain attached to the lung vasculature or independently migrate along the inner walls of vessels. Using fluorescent lineage reporters and flow cytometry, we observed 'waves' of distinct myeloid cell subsets that load differentially and sequentially with this CTC-derived material. Many of these tumour-ingesting myeloid cells collectively accumulated in the lung interstitium along with the successful metastatic cells and, as previously understood, promote the development of successful metastases from surviving tumour cells. Although the numbers of these cells rise globally in the lung with metastatic exposure and ingesting myeloid cells undergo phenotypic changes associated with microparticle ingestion, a consistently sparse population of resident conventional dendritic cells, among the last cells to interact with CTCs, confer anti-metastatic protection. This work reveals that CTC fragmentation generates immune-interacting intermediates, and defines a competitive relationship between phagocyte populations for tumour loading during metastatic cell seeding.


Assuntos
Movimento Celular , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Pulmão/imunologia , Pulmão/patologia , Metástase Neoplásica/imunologia , Metástase Neoplásica/patologia , Animais , Capilares/patologia , Linhagem Celular Tumoral , Linhagem da Célula , Células Dendríticas/citologia , Células Dendríticas/imunologia , Feminino , Genes Reporter/genética , Humanos , Pulmão/irrigação sanguínea , Pulmão/citologia , Neoplasias Pulmonares/patologia , Masculino , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Microscopia Confocal , Células Mieloides/citologia , Células Neoplásicas Circulantes/patologia
13.
Nature ; 517(7536): 621-5, 2015 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-25533958

RESUMO

Broadly, tissue regeneration is achieved in two ways: by proliferation of common differentiated cells and/or by deployment of specialized stem/progenitor cells. Which of these pathways applies is both organ- and injury-specific. Current models in the lung posit that epithelial repair can be attributed to cells expressing mature lineage markers. By contrast, here we define the regenerative role of previously uncharacterized, rare lineage-negative epithelial stem/progenitor (LNEP) cells present within normal distal lung. Quiescent LNEPs activate a ΔNp63 (a p63 splice variant) and cytokeratin 5 remodelling program after influenza or bleomycin injury in mice. Activated cells proliferate and migrate widely to occupy heavily injured areas depleted of mature lineages, at which point they differentiate towards mature epithelium. Lineage tracing revealed scant contribution of pre-existing mature epithelial cells in such repair, whereas orthotopic transplantation of LNEPs, isolated by a definitive surface profile identified through single-cell sequencing, directly demonstrated the proliferative capacity and multipotency of this population. LNEPs require Notch signalling to activate the ΔNp63 and cytokeratin 5 program, and subsequent Notch blockade promotes an alveolar cell fate. Persistent Notch signalling after injury led to parenchymal 'micro-honeycombing' (alveolar cysts), indicative of failed regeneration. Lungs from patients with fibrosis show analogous honeycomb cysts with evidence of hyperactive Notch signalling. Our findings indicate that distinct stem/progenitor cell pools repopulate injured tissue depending on the extent of the injury, and the outcomes of regeneration or fibrosis may depend in part on the dynamics of LNEP Notch signalling.


Assuntos
Células Epiteliais/citologia , Células Epiteliais/patologia , Lesão Pulmonar/patologia , Pulmão/citologia , Pulmão/patologia , Reepitelização , Células-Tronco/citologia , Animais , Bleomicina , Linhagem da Célula , Proliferação de Células , Separação Celular , Cistos/metabolismo , Cistos/patologia , Células Epiteliais/metabolismo , Feminino , Humanos , Queratina-5/metabolismo , Pulmão/fisiologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/virologia , Masculino , Camundongos , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Transplante de Células-Tronco , Células-Tronco/metabolismo , Transativadores/genética , Transativadores/metabolismo
14.
Arch Toxicol ; 95(2): 693-701, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33084937

RESUMO

Formaldehyde (FA), an economically important and ubiquitous chemical, has been classified as a human carcinogen and myeloid leukemogen. However, the underlying mechanisms of leukemogenesis remain unclear. Unlike many classical leukemogens that damage hematopoietic stem/progenitor cells (HSC/HPC) directly in the bone marrow, FA-as the smallest, most reactive aldehyde-is thought to be incapable of reaching the bone marrow through inhalation exposure. A recent breakthrough study discovered that mouse lung contains functional HSC/HPC that can produce blood cells and travel bi-directionally between the lung and bone marrow, while another early study reported the presence of HSC/HPC in rat nose. Based on these findings, we hypothesized that FA inhalation could induce toxicity in HSC/HPC present in mouse lung and/or nose rather than in the bone marrow. To test this hypothesis, we adapted a commercially available protocol for culturing burst-forming unit-erythroid (BFU-E) and colony-forming unit-granulocyte, macrophage (CFU-GM) colonies from bone marrow and spleen to also enable culture of these colonies from mouse lung and nose, a novel application of this assay. We reported that in vivo exposure to FA at 3 mg/m3 or ex vivo exposure up to 400 µM FA decreased the formation of both colony types from mouse lung and nose as well as from bone marrow and spleen. These findings, to the best of our knowledge, are the first empirically to show that FA exposure can damage mouse pulmonary and olfactory HSC/HPC and provide potential biological plausibility for the induction of leukemia at the sites of entry rather than the bone marrow.


Assuntos
Formaldeído/toxicidade , Células-Tronco Hematopoéticas/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Nariz/efeitos dos fármacos , Animais , Células da Medula Óssea/efeitos dos fármacos , Carcinógenos/toxicidade , Células Cultivadas , Exposição por Inalação , Leucemia/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Organismos Livres de Patógenos Específicos , Baço/efeitos dos fármacos
15.
Am J Respir Cell Mol Biol ; 62(3): 364-372, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31647878

RESUMO

The immune system is designed to robustly respond to pathogenic stimuli but to be tolerant to endogenous ligands to not trigger autoimmunity. Here, we studied an endogenous damage-associated molecular pattern, mitochondrial DNA (mtDNA), during primary graft dysfunction (PGD) after lung transplantation. We hypothesized that cell-free mtDNA released during lung ischemia-reperfusion triggers neutrophil extracellular trap (NET) formation via TLR9 signaling. We found that mtDNA increases in the BAL fluid of experimental PGD (prolonged cold ischemia followed by orthotopic lung transplantation) and not in control transplants with minimal warm ischemia. The adoptive transfer of mtDNA into the minimal warm ischemia graft immediately before lung anastomosis induces NET formation and lung injury. TLR9 deficiency in neutrophils prevents mtDNA-induced NETs, and TLR9 deficiency in either the lung donor or recipient decreases NET formation and lung injury in the PGD model. Compared with human lung transplant recipients without PGD, severe PGD was associated with high levels of BAL mtDNA and NETs, with evidence of relative deficiency in DNaseI. We conclude that mtDNA released during lung ischemia-reperfusion triggers TLR9-dependent NET formation and drives lung injury. In PGD, DNaseI therapy has a potential dual benefit of neutralizing a major NET trigger (mtDNA) in addition to dismantling pathogenic NETs.


Assuntos
Isquemia Fria/efeitos adversos , DNA Mitocondrial/farmacologia , Armadilhas Extracelulares/metabolismo , Neutrófilos/efeitos dos fármacos , Disfunção Primária do Enxerto/imunologia , Receptor Toll-Like 9/fisiologia , Lesão Pulmonar Aguda/etiologia , Animais , Líquido da Lavagem Broncoalveolar/citologia , Citrulinação , DNA Mitocondrial/administração & dosagem , Desoxirribonuclease I/metabolismo , Humanos , Transplante de Pulmão , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/imunologia , Disfunção Primária do Enxerto/metabolismo , Proteína-Arginina Desiminase do Tipo 4/deficiência , Proteína-Arginina Desiminase do Tipo 4/fisiologia , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/metabolismo , Organismos Livres de Patógenos Específicos , Receptor Toll-Like 9/deficiência , Isquemia Quente/efeitos adversos
16.
Physiology (Bethesda) ; 34(6): 392-401, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31577166

RESUMO

Megakaryocytes are normal cellular components of the blood returning to the heart and entering the lungs, and historical data has pointed to a role of the lungs in platelet production. Recent studies using intravital microscopy have demonstrated that platelet release occurs in the lung from bone marrow megakaryocytes that embolize into the lung circulation.


Assuntos
Plaquetas/fisiologia , Pulmão/fisiologia , Animais , Medula Óssea/fisiologia , Humanos , Megacariócitos/fisiologia
17.
Cell Immunol ; 350: 103862, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-30336937

RESUMO

The lung represents a unique immune environment. The primary function of the lung is to enable gas exchange by facilitating the transfer of oxygen into and carbon dioxide out of the blood. However, as a direct byproduct of this process the lung is also constantly exposed to particles, allergens, and pathogens alongside air itself. Due to this, the pulmonary immune system exists in a fine balance between quiescence and inflammation, deviations from which can lead to a failure in respiratory function. A rich history exists attempting to define the critical features of lung immunity, and most recently advances in intravital microscopy have enabled the visualization of intercellular immune dynamics in both steady-state and a variety of disease conditions. In this review, we will summarize a variety of approaches to intravital lung imaging as well as how its application has advanced our understanding of normal lung function as well as disease states such as pulmonary metastasis, asthma, and lung injury.


Assuntos
Microscopia Intravital/métodos , Pulmão/diagnóstico por imagem , Pulmão/imunologia , Alérgenos/imunologia , Asma/imunologia , Humanos , Sistema Imunitário/imunologia , Inflamação/patologia , Microscopia de Fluorescência por Excitação Multifotônica/métodos
18.
Transfusion ; 60(11): 2548-2556, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32905629

RESUMO

BACKGROUND: Consensus definitions for transfusion-related acute lung injury (TRALI) and transfusion-associated circulatory overload (TACO) have recently been revised; however, pulmonary transfusion reactions remain difficult to diagnose. We hypothesized that N-terminal pro-brain natriuretic peptide (NT-proBNP) levels could have utility in the identification and classification of pulmonary transfusion reactions. STUDY DESIGN AND METHODS: We performed a secondary analysis of a case-control study of pulmonary transfusion reactions at four academic hospitals. We evaluated clinical data and measured NT-proBNP levels prior to and following transfusion in patients with TACO (n = 160), transfused acute respiratory distress syndrome (ARDS) [n = 51], TRALI [n = 12], TACO/TRALI [n = 7], and controls [n = 335]. We used Wilcoxon Rank-Sum tests to compare NT-proBNP levels, and classification and regression tree (CART) algorithms to produce a ranking of covariates in order of relative importance for differentiating TACO from transfused controls. RESULTS: Pre-transfusion NT-proBNP levels were elevated in cases of transfused ARDS and TACO (both P < .001) but not TRALI (P = .31) or TACO/TRALI (P = .23) compared to transfused controls. Pre-transfusion NT-proBNP levels were higher in cases of transfused ARDS or TRALI with a diagnosis of sepsis compared to those without (P < .05 for both). CART analyses resulted in similar differentiation of patients with TACO from transfused controls for models utilizing either NT-proBNP levels (AUC 0.83) or echocardiogram results (AUC 0.80). CONCLUSIONS: NT-proBNP levels may have utility in the classification of pulmonary transfusion reactions. Prospective studies are needed to test the predictive utility of pre-transfusion NT-proBNP in conjunction with other clinical factors in identifying patients at risk of pulmonary transfusion reactions.


Assuntos
Peptídeo Natriurético Encefálico/sangue , Fragmentos de Peptídeos/sangue , Síndrome do Desconforto Respiratório , Lesão Pulmonar Aguda Relacionada à Transfusão , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Síndrome do Desconforto Respiratório/sangue , Síndrome do Desconforto Respiratório/classificação , Lesão Pulmonar Aguda Relacionada à Transfusão/sangue , Lesão Pulmonar Aguda Relacionada à Transfusão/classificação
19.
Am J Respir Crit Care Med ; 199(9): 1076-1085, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30888839

RESUMO

Rationale: Extracellular DNA (eDNA) and neutrophil extracellular traps (NETs) are implicated in multiple inflammatory diseases. NETs mediate inflammasome activation and IL-1ß secretion from monocytes and cause airway epithelial cell injury, but the role of eDNA, NETs, and IL-1ß in asthma is uncertain. Objectives: To characterize the role of activated neutrophils in severe asthma through measurement of NETs and inflammasome activation. Methods: We measured sputum eDNA in induced sputum from 399 patients with asthma in the Severe Asthma Research Program-3 and in 94 healthy control subjects. We subdivided subjects with asthma into eDNA-low and -high subgroups to compare outcomes of asthma severity and of neutrophil and inflammasome activation. We also examined if NETs cause airway epithelial cell damage that can be prevented by DNase. Measurements and Main Results: We found that 13% of the Severe Asthma Research Program-3 cohort is "eDNA-high," as defined by sputum eDNA concentrations above the upper 95th percentile value in health. Compared with eDNA-low patients with asthma, eDNA-high patients had lower Asthma Control Test scores, frequent history of chronic mucus hypersecretion, and frequent use of oral corticosteroids for maintenance of asthma control (all P values <0.05). Sputum eDNA in asthma was associated with airway neutrophilic inflammation, increases in soluble NET components, and increases in caspase 1 activity and IL-1ß (all P values <0.001). In in vitro studies, NETs caused cytotoxicity in airway epithelial cells that was prevented by disruption of NETs with DNase. Conclusions: High extracellular DNA concentrations in sputum mark a subset of patients with more severe asthma who have NETs and markers of inflammasome activation in their airways.


Assuntos
Asma/fisiopatologia , DNA/metabolismo , Armadilhas Extracelulares/fisiologia , Inflamassomos/fisiologia , Doença Aguda , Adulto , Asma/imunologia , Asma/metabolismo , Western Blotting , Estudos de Casos e Controles , Feminino , Glucosefosfato Desidrogenase/metabolismo , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Neutrófilos/fisiologia
20.
J Bacteriol ; 201(22)2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31481541

RESUMO

Conditionally essential (CE) genes are required by pathogenic bacteria to establish and maintain infections. CE genes encode virulence factors, such as secretion systems and effector proteins, as well as biosynthetic enzymes that produce metabolites not found in the host environment. Due to their outsized importance in pathogenesis, CE gene products are attractive targets for the next generation of antimicrobials. However, the precise manipulation of CE gene expression in the context of infection is technically challenging, limiting our ability to understand the roles of CE genes in pathogenesis and accordingly design effective inhibitors. We previously developed a suite of CRISPR interference-based gene knockdown tools that are transferred by conjugation and stably integrate into bacterial genomes that we call Mobile-CRISPRi. Here, we show the efficacy of Mobile-CRISPRi in controlling CE gene expression in an animal infection model. We optimize Mobile-CRISPRi in Pseudomonas aeruginosa for use in a murine model of pneumonia by tuning the expression of CRISPRi components to avoid nonspecific toxicity. As a proof of principle, we demonstrate that knock down of a CE gene encoding the type III secretion system (T3SS) activator ExsA blocks effector protein secretion in culture and attenuates virulence in mice. We anticipate that Mobile-CRISPRi will be a valuable tool to probe the function of CE genes across many bacterial species and pathogenesis models.IMPORTANCE Antibiotic resistance is a growing threat to global health. To optimize the use of our existing antibiotics and identify new targets for future inhibitors, understanding the fundamental drivers of bacterial growth in the context of the host immune response is paramount. Historically, these genetic drivers have been difficult to manipulate precisely, as they are requisite for pathogen survival. Here, we provide the first application of Mobile-CRISPRi to study conditionally essential virulence genes in mouse models of lung infection through partial gene perturbation. We envision the use of Mobile-CRISPRi in future pathogenesis models and antibiotic target discovery efforts.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes/métodos , Pneumonia Bacteriana/microbiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/patogenicidade , Animais , Proteína 9 Associada à CRISPR , Técnicas de Silenciamento de Genes , Genes Bacterianos , Immunoblotting , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia Bacteriana/metabolismo , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sistemas de Secreção Tipo III/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA