Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Lung ; 198(6): 909-916, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33106891

RESUMO

BACKGROUND: Lung inflammation is associated with many respiratory conditions. Consequently, anti-inflammatory medications, like glucocorticoids, have become mainstay intrapulmonary therapeutics. However, their effectiveness for treating inflammation occurring in the alveolar regions of the lung is limited by suboptimal delivery. To improve the pulmonary distribution of glucocorticoids, such as budesonide to distal regions of the lung, exogenous surfactant has been proposed as an ideal delivery vehicle for such therapies. It was therefore hypothesized that fortifying an exogenous surfactant (BLES) with budesonide would enhance efficacy for treating pulmonary inflammation in vivo. METHODS: An intratracheal instillation of heat-killed bacteria was used to elicit an inflammatory response in the lungs of male and female rats. Thirty minutes after this initial instillation, either budesonide or BLES combined with budesonide was administered intratracheally. To evaluate the efficacy of surfactant delivery, various markers of inflammation were measured in the bronchoalveolar lavage and lung tissue. RESULTS: Although budesonide exhibited anti-inflammatory effects when administered alone, delivery with BLES enhanced those effects by lowering the lavage neutrophil counts and myeloperoxidase activity in lung tissue. Combining budesonide with BLES was also shown to reduce several other pro-inflammatory mediators. These results were shown across both sexes, with no observed sex differences. CONCLUSION: Based on these findings, it was concluded that exogenous surfactant can enhance the delivery and efficacy of budesonide in vivo.


Assuntos
Produtos Biológicos/administração & dosagem , Budesonida/administração & dosagem , Glucocorticoides/administração & dosagem , Pneumonia/tratamento farmacológico , Surfactantes Pulmonares/administração & dosagem , Animais , Modelos Animais de Doenças , Feminino , Masculino , Veículos Farmacêuticos , Pneumonia/etiologia , Ratos , Ratos Wistar
3.
Infect Immun ; 85(12)2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28947647

RESUMO

The development of antibiotic resistance by Pseudomonas aeruginosa is a major concern in the treatment of bacterial pneumonia. In the search for novel anti-infective therapies, the chicken-derived peptide cathelicidin-2 (CATH-2) has emerged as a potential candidate, with strong broad-spectrum antimicrobial activity and the ability to limit inflammation by inhibiting Toll-like receptor 2 (TLR2) and TLR4 activation. However, as it is unknown how CATH-2 affects inflammation in vivo, we investigated how CATH-2-mediated killing of P. aeruginosa affects lung inflammation in a murine model. First, murine macrophages were used to determine whether CATH-2-mediated killing of P. aeruginosa reduced proinflammatory cytokine production in vitro Next, a murine lung model was used to analyze how CATH-2-mediated killing of P. aeruginosa affects neutrophil and macrophage recruitment as well as cytokine/chemokine production in the lung. Our results show that CATH-2 kills P. aeruginosa in an immunogenically silent manner both in vitro and in vivo Treatment with CATH-2-killed P. aeruginosa showed reduced neutrophil recruitment to the lung as well as inhibition of cytokine and chemokine production, compared to treatment with heat- or gentamicin-killed bacteria. Together, these results show the potential for CATH-2 as a dual-activity antibiotic in bacterial pneumonia, which can both kill P. aeruginosa and prevent excessive inflammation.


Assuntos
Peptídeos Catiônicos Antimicrobianos/imunologia , Inflamação/prevenção & controle , Pulmão/microbiologia , Pneumonia Bacteriana/imunologia , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/efeitos dos fármacos , Animais , Peptídeos Catiônicos Antimicrobianos/farmacologia , Linhagem Celular , Quimiocinas/imunologia , Galinhas/imunologia , Citocinas/imunologia , Modelos Animais de Doenças , Imunidade Inata , Inflamação/imunologia , Pulmão/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos , Pneumonia Bacteriana/prevenção & controle , Infecções por Pseudomonas/veterinária
4.
Am J Physiol Lung Cell Mol Physiol ; 313(3): L524-L533, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28546153

RESUMO

Alterations to the pulmonary surfactant system have been observed consistently in ventilation-induced lung injury (VILI) including composition changes and impairments in the surface tension reducing ability of the isolated extracellular surfactant. However, there is limited information about the effects of VILI on the intracellular form of surfactant, the lamellar body. It is hypothesized that VILI leads to alterations of lamellar body numbers and function. To test this hypothesis, rats were randomized to one of three groups, nonventilated controls, control ventilation, and high tidal volume ventilation (VILI). Following physiological assessment to confirm lung injury, isolated lamellar bodies were tested for surfactant function on a constrained sessile drop surfactometer. A separate cohort of animals was used to fix the lungs followed by examination of lamellar body numbers and morphology using transmission electron microscopy. The results showed an impaired ability of reducing surface tension for the lamellar bodies isolated from the VILI group as compared with the two other groups. The morphological assessment revealed that the number, and the relative area covered by, lamellar bodies were significantly decreased in animals with VILI animals as compared with the other groups. It is concluded that VILI causes significant alterations to lamellar bodies. It is speculated that increased secretion causes a depletion of lamellar bodies that cannot be compensated by de novo synthesis of surfactant in these injured lungs.


Assuntos
Lisossomos/patologia , Lesão Pulmonar Induzida por Ventilação Mecânica/patologia , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/ultraestrutura , Animais , Colesterol/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pulmão/fisiopatologia , Lisossomos/efeitos dos fármacos , Lisossomos/ultraestrutura , Masculino , Oxigênio/metabolismo , Fosfolipídeos/metabolismo , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , Surfactantes Pulmonares/farmacologia , Ratos Sprague-Dawley , Tensão Superficial/efeitos dos fármacos , Lesão Pulmonar Induzida por Ventilação Mecânica/fisiopatologia
5.
Cell Tissue Res ; 367(3): 495-509, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27796509

RESUMO

Acute respiratory distress syndrome (ARDS) is a disease with a variety of causes and is defined by severe hypoxemia. Whereas ARDS carries a mortality of approximately 30 %, patients that survive may ultimately regain near normal pulmonary physiology. The critical pathophysiological processes in ARDS are alveolar barrier dysfunction and overwhelming inflammation. This encompasses damage to the epithelial and endothelial layers, thickening of the interstitial matrix, edema with inactivation of pulmonary surfactant at the alveolar surface and marked inflammation mediated by infiltrating neutrophils and pro-inflammatory macrophages. For patients that survive the disease, these are the critical processes that require repair and remodeling to allow for the recovery of ARDS. As such, the current review focuses on the experimental studies that have begun to elucidate the mechanisms involved in restoring the alveolar barrier following injury.


Assuntos
Lesão Pulmonar Aguda/fisiopatologia , Pulmão/fisiopatologia , Lesão Pulmonar Aguda/patologia , Lesão Pulmonar Aguda/terapia , Animais , Humanos , Pulmão/patologia , Modelos Biológicos , Regeneração , Resultado do Tratamento
6.
Biochim Biophys Acta ; 1848(3): 813-20, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25522687

RESUMO

The ability of pulmonary surfactant to reduce surface tension at the alveolar surface is impaired in various lung diseases. Recent animal studies indicate that elevated levels of cholesterol within surfactant may contribute to its inhibition. It was hypothesized that elevated cholesterol levels within surfactant inhibit human surfactant biophysical function and that these effects can be reversed by surfactant protein A (SP-A). The initial experiment examined the function of surfactant from mechanically ventilated trauma patients in the presence and absence of a cholesterol sequestering agent, methyl-ß-cyclodextrin. The results demonstrated improved surface activity when cholesterol was sequestered in vitro using a captive bubble surfactometer (CBS). These results were explored further by reconstitution of surfactant with various concentrations of cholesterol with and without SP-A, and testing of the functionality of these samples in vitro with the CBS and in vivo using surfactant depleted rats. Overall, the results consistently demonstrated that surfactant function was inhibited by levels of cholesterol of 10% (w/w phospholipid) but this inhibition was mitigated by the presence of SP-A. It is concluded that cholesterol-induced surfactant inhibition can actively contribute to physiological impairment of the lungs in mechanically ventilated patients and that SP-A levels may be important to maintain surfactant function in the presence of high cholesterol within surfactant.


Assuntos
Colesterol/metabolismo , Proteína A Associada a Surfactante Pulmonar/metabolismo , Respiração Artificial/métodos , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Colesterol/farmacologia , Feminino , Humanos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/fisiopatologia , Doenças Pulmonares Intersticiais/metabolismo , Doenças Pulmonares Intersticiais/fisiopatologia , Masculino , Microscopia de Força Atômica , Pessoa de Meia-Idade , Oxigênio/sangue , Fosfolipídeos/metabolismo , Fosfolipídeos/farmacologia , Pressão , Proteína A Associada a Surfactante Pulmonar/farmacologia , Surfactantes Pulmonares/metabolismo , Surfactantes Pulmonares/farmacologia , Ratos , Tensão Superficial/efeitos dos fármacos , Adulto Jovem , beta-Ciclodextrinas/metabolismo , beta-Ciclodextrinas/farmacologia
7.
Exp Lung Res ; 42(7): 365-379, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27676418

RESUMO

BACKGROUND: The acute respiratory distress syndrome (ARDS) is a complex pulmonary disorder in which the local release of cytokines and chemokines appears central to the pathophysiology. OBJECTIVE: Based on the known role of matrix metalloproteinase-3 (MMP3) in inflammatory processes, the objective was to examine the role of MMP3 in the pathogenesis of ARDS through the modulation of pulmonary inflammation. MATERIALS AND METHODS: Female and male, wild type (MMP3+/+) and knock out (MMP3-/-) mice were exposed to two, clinically relevant models of ARDS including (i) lipopolysaccharide (LPS)-induced lung injury, and (ii) hydrochloric acid-induced lung injury. Parameters of lung injury and inflammation were assessed through measurements in lung lavage including total protein content, inflammatory cell influx, and concentrations of mediators such as TNF-α, IL-6, G-CSF, CXCL1, CXCL2, and CCL2. Lung histology and compliance were also evaluated in the LPS model of injury. RESULTS: Following intra-tracheal LPS instillation, all mice developed lung injury, as measured by an increase in lavage neutrophils, and decrease in lung compliance, with no overall effect of genotype observed. Increased concentrations of lavage inflammatory cytokines and chemokines were also observed following LPS injury, however, LPS-instilled female MMP3-/- mice had lower levels of inflammatory mediators compared to LPS-instilled female MMP3+/+ mice. This effect of the genotype was not observed in male mice. Similar findings, including the MMP3-related sex differences, were also observed after acid-induced lung injury. CONCLUSION: MMP3 contributes to the pathogenesis of ARDS, by affecting the pulmonary inflammatory response in female mice in relevant models of lung injury.


Assuntos
Metaloproteinase 3 da Matriz/farmacologia , Pneumonia/induzido quimicamente , Síndrome do Desconforto Respiratório/etiologia , Lesão Pulmonar Aguda/induzido quimicamente , Animais , Feminino , Humanos , Ácido Clorídrico/farmacologia , Lipopolissacarídeos/farmacologia , Masculino , Metaloproteinase 3 da Matriz/genética , Camundongos , Fatores Sexuais
8.
Can J Physiol Pharmacol ; 94(6): 682-5, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27096327

RESUMO

The acute respiratory distress syndrome (ARDS) is characterized by arterial hypoxemia accompanied by severe inflammation and alterations to the pulmonary surfactant system. Published data has demonstrated a protective effect of matrix metalloproteinase-3 (Mmp3) deficiency against the inflammatory response associated with ARDS; however, the effect of Mmp3 on physiologic parameters and alterations to surfactant have not been previously studied. It was hypothesized that Mmp3 deficient (Mmp3(-/-)) mice would be protected against lung dysfunction associated with ARDS and maintain a functional pulmonary surfactant system. Wild type (WT) and Mmp3(-/-) mice were subjected to acid-aspiration followed by mechanical ventilation. Mmp3(-/-) mice maintained higher arterial oxygenation compared with WT mice at the completion of ventilation. Significant increase in functional large aggregate surfactant forms were observed in Mmp3(-/-) mice compared with WT mice. These findings further support a role of Mmp3 as an attractive therapeutic target for drug development in the setting of ARDS.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Modelos Animais de Doenças , Metaloproteinase 3 da Matriz/deficiência , Surfactantes Pulmonares/metabolismo , Síndrome do Desconforto Respiratório/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Líquido da Lavagem Broncoalveolar , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Distribuição Aleatória , Síndrome do Desconforto Respiratório/patologia
9.
Antimicrob Agents Chemother ; 59(6): 3075-83, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25753641

RESUMO

Antibiotic-resistant bacterial infections represent an emerging health concern in clinical settings, and a lack of novel developments in the pharmaceutical pipeline is creating a "perfect storm" for multidrug-resistant bacterial infections. Antimicrobial peptides (AMPs) have been suggested as future therapeutics for these drug-resistant bacteria, since they have potent broad-spectrum activity, with little development of resistance. Due to the unique structure of the lung, bacterial pneumonia has the additional problem of delivering antimicrobials to the site of infection. One potential solution is coadministration of AMPs with exogenous surfactant, allowing for distribution of the peptides to distal airways and opening of collapsed lung regions. The objective of this study was to test various surfactant-AMP mixtures with regard to maintaining pulmonary surfactant biophysical properties and bactericidal functions. We compared the properties of four AMPs (CATH-1, CATH-2, CRAMP, and LL-37) suspended in bovine lipid-extract surfactant (BLES) by assessing surfactant-AMP mixture biophysical and antimicrobial functions. Antimicrobial activity was tested against methillicin-resistant Staphylococcus aureus and Pseudomonas aeruginosa. All AMP/surfactant mixtures exhibited an increase of spreading compared to a BLES control. BLES+CATH-2 mixtures had no significantly different minimum surface tension versus the BLES control. Compared to the other cathelicidins, CATH-2 retained the most bactericidal activity in the presence of BLES. The BLES+CATH-2 mixture appears to be an optimal surfactant-AMP mixture based on in vitro assays. Future directions involve investigating the potential of this mixture in animal models of bacterial pneumonia.


Assuntos
Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Pneumonia Bacteriana/tratamento farmacológico , Pneumonia Bacteriana/microbiologia , Surfactantes Pulmonares/farmacologia , Surfactantes Pulmonares/uso terapêutico , Animais , Peptídeos Catiônicos Antimicrobianos/farmacologia , Bovinos , Masculino , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Camundongos , Pseudomonas aeruginosa/efeitos dos fármacos , Staphylococcus aureus/efeitos dos fármacos
10.
Am J Pathol ; 184(5): 1489-502, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24650563

RESUMO

Fibrotic lung diseases represent a diverse group of progressive and often fatal disorders with limited treatment options. Although the pathogenesis of these conditions remains incompletely understood, receptor type protein tyrosine phosphatase α (PTP-α encoded by PTPRA) has emerged as a key regulator of fibroblast signaling. We previously reported that PTP-α regulates cellular responses to cytokines and growth factors through integrin-mediated signaling and that PTP-α promotes fibroblast expression of matrix metalloproteinase 3, a matrix-degrading proteinase linked to pulmonary fibrosis. Here, we sought to determine more directly the role of PTP-α in pulmonary fibrosis. Mice genetically deficient in PTP-α (Ptpra(-/-)) were protected from pulmonary fibrosis induced by intratracheal bleomycin, with minimal alterations in the early inflammatory response or production of TGF-ß. Ptpra(-/-) mice were also protected from pulmonary fibrosis induced by adenoviral-mediated expression of active TGF-ß1. In reciprocal bone marrow chimera experiments, the protective phenotype tracked with lung parenchymal cells but not bone marrow-derived cells. Because fibroblasts are key contributors to tissue fibrosis, we compared profibrotic responses in wild-type and Ptpra(-/-) mouse embryonic and lung fibroblasts. Ptpra(-/-) fibroblasts exhibited hyporesponsiveness to TGF-ß, manifested by diminished expression of αSMA, EDA-fibronectin, collagen 1A, and CTGF. Ptpra(-/-) fibroblasts exhibited markedly attenuated TGF-ß-induced Smad2/3 transcriptional activity. We conclude that PTP-α promotes profibrotic signaling pathways in fibroblasts through control of cellular responsiveness to TGF-ß.


Assuntos
Fibroblastos/patologia , Pulmão/patologia , Fibrose Pulmonar/patologia , Proteínas Tirosina Fosfatases Classe 4 Semelhantes a Receptores/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Adenoviridae , Animais , Bleomicina , Citocinas/biossíntese , Deleção de Genes , Genes Reporter , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Pneumonia/complicações , Pneumonia/patologia , Fibrose Pulmonar/complicações , Fibrose Pulmonar/prevenção & controle , Proteínas Tirosina Fosfatases Classe 4 Semelhantes a Receptores/deficiência , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas Smad/metabolismo , Transcrição Gênica
12.
Respiration ; 87(5): 416-27, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24662316

RESUMO

BACKGROUND: Apolipoprotein E (apoE) has been shown to play a pivotal role in the development of cardiovascular disease, attributable to its function in lipid trafficking and immune modulating properties; however, its role in modulating inflammation in the setting of acute lung injury (ALI) is unknown. OBJECTIVE: To determine whether apoE-deficient mice (apoE-/-) are more susceptible to ALI compared to wild-type (WT) animals. METHODS: Two independent models of ALI were employed. Firstly, WT and apoE-/- mice were randomized to acid aspiration (50 µl of 0.1 N hydrochloric acid) followed by 4 h of mechanical ventilation. Secondly, WT and apoE-/- mice were randomized to 72 h of hyperoxia exposure or room air. Thereafter, the intrinsic responses of WT and apoE-/- mice were assessed using the isolated perfused mouse lung (IPML) setup. Finally, based on elevated levels of oxidized low-density lipoprotein (oxLDL) in apoE-/-, the effect of oxLDL on lung endothelial permeability and inflammation was assessed. RESULTS: In both in vivo models, apoE-/- mice demonstrated greater increases in lung lavage protein levels, neutrophil counts, and cytokine expression (p < 0.05) compared to WT mice. Experiments utilizing the IPML setup demonstrated no differences in intrinsic lung responses to injury between apoE-/- and WT mice, suggesting the presence of a circulating factor as being responsible for the in vivo observations. Finally, the exposure of lung endothelial cells to oxLDL resulted in increased monolayer permeability and IL-6 release compared to native (nonoxidized) LDL. CONCLUSIONS: Our findings demonstrate a susceptibility of apoE-/- animals to ALI that may occur, in part, due to elevated levels of oxLDL.


Assuntos
Lesão Pulmonar Aguda/genética , Apolipoproteínas E/genética , Lipoproteínas LDL/metabolismo , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/metabolismo , Animais , Modelos Animais de Doenças , Predisposição Genética para Doença , Ácido Clorídrico/toxicidade , Inflamação , Interleucina-6/metabolismo , Lipoproteínas LDL/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Camundongos , Camundongos Knockout , Permeabilidade/efeitos dos fármacos , Respiração Artificial/efeitos adversos , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo
13.
Chest ; 164(1): 27-38, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36781102

RESUMO

BACKGROUND: We previously showed in patients with poorly controlled eosinophilic asthma that a single dose of benralizumab resulted in significantly improved Asthma Control Questionnaire (ACQ-6) score and 129Xe MRI ventilation defect percent (VDP) 28 days postinjection, and 129Xe MRI VDP and CT airway mucus occlusions were shown to independently predict this early ACQ-6 response to benralizumab. RESEARCH QUESTION: Do early VDP responses at 28 days persist, and do FEV1, fractional exhaled nitric oxide, and mucus plug score improve during a 2.5 year treatment period? STUDY DESIGN AND METHODS: Participants with poorly controlled eosinophilic asthma completed spirometry, ACQ-6, and MRI, 28 days, 1 year, and 2.5 years after initiation of treatment with benralizumab; chest CT was acquired at enrollment and 2.5 years later. RESULTS: Of 29 participants evaluated at 28 days post-benralizumab, 16 participants returned for follow-up while on therapy at 1 year, and 13 participants were evaluable while on therapy at 2.5 years post-benralizumab initiation. As compared with 28 days post-benralizumab, ACQ-6 score (2.0 ± 1.4) significantly improved after 1 year (0.5 ± 0.6, P = .02; 95% CI, 0.1-1.1) and 2.5 years (0.5 ± 0.5, P = .03; 95% CI, 0.1-1.1). The mean VDP change at 2.5 years (-4% ± 3%) was greater than the minimal clinically important difference, but not significantly different from VDP measured 28 days post-benralizumab. Mucus score (3 ± 4) was significantly improved at 2.5 years (1 ± 1, P = .03; 95% CI, 0.3-5.5). In six of eight participants with previous occlusions, mucus plugs vanished or substantially diminished 2.5 years later. VDP (P < .001) and mucus score (P < .001) measured at baseline, but not fractional exhaled nitric oxide or FEV1, independently predicted ACQ-6 score after 2.5 years. INTERPRETATION: In poorly controlled eosinophilic asthma, early MRI VDP responses at 28 days post-benralizumab persisted 2.5 years later, alongside significantly improved mucus scores and asthma control.


Assuntos
Obstrução das Vias Respiratórias , Antiasmáticos , Asma , Eosinofilia Pulmonar , Humanos , Óxido Nítrico , Asma/diagnóstico por imagem , Asma/tratamento farmacológico , Eosinofilia Pulmonar/tratamento farmacológico , Muco , Imageamento por Ressonância Magnética/métodos , Tomografia Computadorizada por Raios X , Antiasmáticos/uso terapêutico
14.
Acad Radiol ; 30(12): 3114-3123, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37032278

RESUMO

RATIONALE AND OBJECTIVES: The minimal clinically important difference (MCID) and upper limit of normal (ULN) for MRI ventilation defect percent (VDP) were previously reported for hyperpolarized 3He gas MRI. Hyperpolarized 129Xe VDP is more sensitive to airway dysfunction than 3He, therefore the objective of this study was to determine the ULN and MCID for 129Xe MRI VDP in healthy and asthma participants. MATERIALS AND METHODS: We retrospectively evaluated healthy and asthma participants who underwent spirometry and 129XeMRI on a single visit; participants with asthma completed the asthma control questionnaire (ACQ-7). The MCID was estimated using distribution- (smallest detectable difference [SDD]) and anchor-based (ACQ-7) methods. Two observers measured VDP (semiautomated k-means-cluster segmentation algorithm) in 10 participants with asthma, five-times each in random order, to determine SDD. The ULN was estimated based on the 95% confidence interval of the relationships between VDP and age. RESULTS: Mean VDP was 1.6 ± 1.2% for healthy (n = 27) and 13.7 ± 12.9% for asthma participants (n = 55). ACQ-7 and VDP were correlated (r = .37, p = .006; VDP = 3.5·ACQ + 4.9). The anchor-based MCID was 1.75% while the mean SDD and distribution-based MCID was 2.25%. VDP was correlated with age for healthy participants (p = .56, p =.003; VDP = .04·Age-.01). The ULN for all healthy participants was 2.0%. By age tertiles, the ULN was 1.3% ages 18-39 years, 2.5% for 40-59 years and 3.8% for 60-79 years. CONCLUSION: The 129Xe MRI VDP MCID was estimated in participants with asthma; the ULN was estimated in healthy participants across a range of ages, both of which provide a way to interpret VDP measurements in clinical investigations.


Assuntos
Asma , Pulmão , Humanos , Lactente , Adolescente , Adulto Jovem , Adulto , Estudos Retrospectivos , Diferença Mínima Clinicamente Importante , Asma/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos
15.
J Otolaryngol Head Neck Surg ; 52(1): 30, 2023 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-37095527

RESUMO

BACKGROUND: Chronic rhinosinusitis with nasal polyposis (CRSwNP) often coexists with lower airway disease. With the overlap between upper and lower airway disease, optimal management of the upper airways is undertaken in conjunction with that of the lower airways. Biologic therapy with targeted activity within the Type 2 inflammatory pathway can improve the clinical signs and symptoms of both upper and lower airway diseases. Knowledge gaps nevertheless exist in how best to approach patient care as a whole. There have been sixteen randomized, double-blind, placebo-controlled trails performed for CRSwNP targeted components of the Type 2 inflammatory pathway, notably interleukin (IL)-4, IL-5 and IL-13, IL- 5R, IL-33, and immunoglobulin (Ig)E. This white paper considers the perspectives of experts in various disciplines such as rhinology, allergy, and respirology across Canada, all of whom have unique and valuable insights to contribute on how to best approach patients with upper airway disease from a multidisciplinary perspective. METHODS: A Delphi Method process was utilized involving three rounds of questionnaires in which the first two were completed individually online and the third was discussed on a virtual platform with all the panelists. A national multidisciplinary expert panel of 34 certified specialists was created, composed of 16 rhinologists, 7 allergists, and 11 respirologists who evaluated the 20 original statements on a scale of 1-9 and provided comments. All ratings were quantitively reviewed by mean, median, mode, range, standard deviation and inter-rater reliability. Consensus was defined by relative interrater reliability measures-kappa coefficient ([Formula: see text]) value > 0.61. RESULTS: After three rounds, a total of 22 statements achieved consensus. This white paper only contains the final agreed upon statements and clear rationale and support for the statements regarding the use of biologics in patients with upper airway disease. CONCLUSION: This white paper provides guidance to Canadian physicians on the use of biologic therapy for the management of upper airway disease from a multidisciplinary perspective, but the medical and surgical regimen should ultimately be individualized to the patient. As more biologics become available and additional trials are published we will provide updated versions of this white paper every few years.


Assuntos
Produtos Biológicos , Pólipos Nasais , Rinite , Sinusite , Humanos , Produtos Biológicos/uso terapêutico , Canadá , Doença Crônica , Consenso , Técnica Delphi , Pólipos Nasais/metabolismo , Reprodutibilidade dos Testes , Rinite/tratamento farmacológico , Sinusite/tratamento farmacológico
16.
Physiology (Bethesda) ; 26(5): 334-47, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22013192

RESUMO

This manuscript will review our current understanding of neutrophilic polymorphonuclear leukocyte (neutrophil) interactions with the endothelium during immune and inflammatory responses, focusing on the molecular mechanisms regulating neutrophil adhesion to and migration through the endothelium in response to infection or tissue injury. This is a complex and dynamic area of research and one that has been the topic of several recent comprehensive reviews to which the interested reader is referred (64, 118, 131). By design, this review will begin with a brief review of some basic aspects of neutrophil biology and endothelial adhesion to provide a foundation. The remainder of the review will focus on selected areas of this complex field, specifically the role of the endothelial glycocalyx in regulating neutrophil adhesion and the mechanisms and consequences of migration of neutrophils between (paracellular) and through (transcellular) endothelial cells during egress from the vasculature.


Assuntos
Comunicação Celular/imunologia , Células Endoteliais/citologia , Células Endoteliais/imunologia , Endotélio/imunologia , Neutrófilos/citologia , Neutrófilos/imunologia , Animais , Adesão Celular/imunologia , Humanos , Imunidade Inata/fisiologia
17.
Am J Pathol ; 179(4): 1733-45, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21871427

RESUMO

Idiopathic pulmonary fibrosis (IPF) may be triggered by epithelial injury that results in aberrant production of growth factors, cytokines, and proteinases, leading to proliferation of myofibroblasts, excess deposition of collagen, and destruction of the lung architecture. The precise mechanisms and key signaling mediators responsible for this aberrant repair process remain unclear. We assessed the importance of matrix metalloproteinase-3 (MMP-3) in the pathogenesis of IPF through i) determination of MMP-3 expression in patients with IPF, ii) in vivo experiments examining the relevance of MMP-3 in experimental models of fibrosis, and iii) in vitro experiments to elucidate possible mechanisms of action. Gene expression analysis, quantitative RT-PCR, and Western blot analysis of explanted human lungs revealed enhanced expression of MMP-3 in IPF, compared with control. Transient adenoviral vector-mediated expression of recombinant MMP-3 in rat lung resulted in accumulation of myofibroblasts and pulmonary fibrosis. Conversely, MMP-3-null mice were protected against bleomycin-induced pulmonary fibrosis. In vitro treatment of cultured lung epithelial cells with purified MMP-3 resulted in activation of the ß-catenin signaling pathway, via cleavage of E-cadherin, and induction of epithelial-mesenchymal transition. These processes were inhibited in bleomycin-treated MMP-3-null mice, as assessed by cytosolic translocation of ß-catenin and cyclin D1 expression. These observations support a novel role for MMP-3 in the pathogenesis of IPF, through activation of ß-catenin signaling and induction of epithelial-mesenchymal transition.


Assuntos
Metaloproteinase 3 da Matriz/metabolismo , Fibrose Pulmonar/enzimologia , Fibrose Pulmonar/patologia , Adenoviridae/genética , Animais , Bleomicina , Caderinas/metabolismo , Ciclina D1/metabolismo , Modelos Animais de Doenças , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal , Feminino , Regulação Enzimológica da Expressão Gênica , Vetores Genéticos/administração & dosagem , Humanos , Pulmão/enzimologia , Pulmão/patologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 3 da Matriz/deficiência , Metaloproteinase 3 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transporte Proteico , Fibrose Pulmonar/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , beta Catenina/metabolismo
18.
Expert Opin Emerg Drugs ; 17(1): 1-4, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22390652

RESUMO

In 2007, Bosma et. al provided a comprehensive review of emerging therapies for the acute respiratory distress syndrome (ARDS), a condition which continues to carry a mortality rate of greater than 30%. Over the past several years, the development of novel and effective therapeutic agents for ARDS remains disappointing, and unfortunately, no recent therapeutic interventions have demonstrated a clear benefit. Herein, the results of several of these early and late phase clinical trials are reviewed, the majority of which address known maladaptive processes that have been deemed critical in ARDS pathophysiology. Based on the ongoing futility of current therapeutic models to yield effective therapies, it is speculated whether or not novel treatment paradigms, which address distinctly different aspects of this disease paradigm, may be warranted.


Assuntos
Drogas em Investigação/uso terapêutico , Insuficiência de Múltiplos Órgãos/etiologia , Síndrome do Desconforto Respiratório/tratamento farmacológico , Medicamentos para o Sistema Respiratório/uso terapêutico , Animais , Humanos
19.
Chest ; 162(3): 520-533, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35283104

RESUMO

BACKGROUND: Patients with eosinophilic asthma often report poor symptomatic control and quality of life. Anti-IL-5 therapy, including anti-IL-5Rα (benralizumab), rapidly depletes eosinophils in the blood and airways and also reduces asthma exacerbations and improves quality of life scores. In patients with severe asthma, eosinophilic inflammation-driven airway mucus occlusions have been measured using thoracic x-ray CT imaging. Pulmonary 129Xe MRI ventilation defect percentage (VDP) also sensitively measures asthma airway dysfunction caused by airway hyperresponsiveness, remodeling, and luminal mucus occlusions. Using 129Xe MRI and CT imaging together, it is feasible to measure both airway luminal occlusions and airway ventilation in relationship to anti-IL-5 therapy to ascertain the direct impact of therapy-induced eosinophil depletion on airway function. RESEARCH QUESTION: Does 129Xe MRI detect airway functional responses to eosinophil depletion after a single benralizumab dose and do airway mucus occlusions mediate this response? STUDY DESIGN AND METHODS: MRI, eosinophil count, spirometry, oscillometry, Asthma Control Questionnaire (ACQ), Asthma Quality of Life Questionnaire (AQLQ), and St. George's Respiratory Questionnaire were completed on day 0 and 28 days after a single 30-mg subcutaneous benralizumab dose. CT scan mucus plugs were scored on day 0, and MRI VDP was quantified on days 0 and 28. RESULTS: Twenty-nine participants (27 with baseline CT imaging) completed day 0 and day 28 visits. On day 28 after a single benralizumab dose, significantly improved blood eosinophil counts, VDP, ACQ 6 scores, AQLQ scores (all P < .001), and peripheral airway resistance (P = .04) were found in all participants. On day 28, significantly improved VDP and ACQ 6 scores also were found in the subgroup of nine participants with five or more mucus plugs, but not in the subgroup (n = 18) with fewer than five mucus plugs. Based on univariate relationships for change in ACQ 6 score, multivariate models were generated and showed that day 0 VDP (P < .001) and day 0 CT scan mucus score (P < .001) were significant variables for change in ACQ 6 score on day 28 after benralizumab injection. INTERPRETATION: 129Xe ventilation significantly improved in participants with uncontrolled asthma and in those with significant mucus plugging after a single dose of benralizumab. TRIAL REGISTRY: ClinicalTrials.gov; No.: NCT03733535; URL: www. CLINICALTRIALS: gov.


Assuntos
Obstrução das Vias Respiratórias , Antiasmáticos , Asma , Eosinofilia Pulmonar , Manuseio das Vias Aéreas , Antiasmáticos/uso terapêutico , Anticorpos Monoclonais Humanizados , Asma/tratamento farmacológico , Eosinófilos , Humanos , Imageamento por Ressonância Magnética/métodos , Muco , Eosinofilia Pulmonar/tratamento farmacológico , Qualidade de Vida , Respiração
20.
Respiration ; 81(4): 333-42, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21311175

RESUMO

BACKGROUND: Overwhelming systemic inflammation has been implicated in the progression of acute lung injury (ALI) leading to multiple organ failure (MOF) and death. Previous studies suggest that mechanical ventilation (MV) may be a key mediator of MOF through an upregulation of the systemic inflammatory response. OBJECTIVES: It was the aim of this study to investigate mechanisms whereby mechanical stress induced by different tidal volumes may contribute to the development of systemic inflammation and maladaptive peripheral organ responses in the setting of ALI. METHODS: An acid aspiration model of ALI was employed in 129X1/SVJ mice through an intratracheal administration of hydrochloric acid followed by MV employing either a low (5 ml/kg) or high (12.5 ml/kg) tidal volume ventilation for 120 min. The isolated perfused mouse lung setup was used to assess the specific contribution of the lung to systemic inflammation during MV. Furthermore, lung perfusate collected over the course of MV was used to assess the effects of lung-derived mediators on activation (expression of a proadhesive phenotype) of liver endothelial cells. RESULTS: High tidal volume MV of acid-injured lungs resulted in greater physiologic and histological indices of lung injury compared to control groups. Additionally, there was an immediate and significant release of multiple inflammatory mediators from the lung into the systemic circulation which resulted in greater levels of mRNA adhesion molecule expression in liver endothelial cells in vitro. CONCLUSIONS: This study suggests that MV, specifically tidal volume strategy, influences the development of MOF through an upregulation of lung-derived systemic inflammation resulting in maladaptive cellular changes in peripheral organs.


Assuntos
Lesão Pulmonar Aguda/fisiopatologia , Insuficiência de Múltiplos Órgãos/etiologia , Respiração Artificial , Volume de Ventilação Pulmonar , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/complicações , Lesão Pulmonar Aguda/patologia , Animais , Líquido da Lavagem Broncoalveolar/química , Moléculas de Adesão Celular/metabolismo , Quimiocinas/análise , Citocinas/análise , Células Endoteliais/metabolismo , Ácido Clorídrico , Inflamação , Pulmão/patologia , Complacência Pulmonar , Masculino , Camundongos , Camundongos Endogâmicos , Insuficiência de Múltiplos Órgãos/fisiopatologia , Respiração Artificial/métodos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa