Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Immunol ; 7(67): eabf7777, 2022 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-34995099

RESUMO

Resident macrophages orchestrate homeostatic, inflammatory, and reparative activities. It is appreciated that different tissues instruct specialized macrophage functions. However, individual tissues contain heterogeneous subpopulations, and how these subpopulations are related is unclear. We asked whether common transcriptional and functional elements could reveal an underlying framework across tissues. Using single-cell RNA sequencing and random forest modeling, we observed that four genes could predict three macrophage subsets that were present in murine heart, liver, lung, kidney, and brain. Parabiotic and genetic fate mapping studies revealed that these core markers predicted three unique life cycles across 17 tissues. TLF+ (expressing TIMD4 and/or LYVE1 and/or FOLR2) macrophages were maintained through self-renewal with minimal monocyte input; CCR2+ (TIMD4−LYVE1−FOLR2−) macrophages were almost entirely replaced by monocytes, and MHC-IIhi macrophages (TIMD4−LYVE1−FOLR2−CCR2−), while receiving modest monocyte contribution, were not continually replaced. Rather, monocyte-derived macrophages contributed to the resident macrophage population until they reached a defined upper limit after which they did not outcompete pre-existing resident macrophages. Developmentally, TLF+ macrophages were first to emerge in the yolk sac and early fetal organs. Fate mapping studies in the mouse and human single-cell RNA sequencing indicated that TLF+ macrophages originated from both yolk sac and fetal monocyte precursors. Furthermore, TLF+ macrophages were the most transcriptionally conserved subset across mouse tissues and between mice and humans, despite organ- and species-specific transcriptional differences. Here, we define the existence of three murine macrophage subpopulations based on common life cycle properties and core gene signatures and provide a common starting point to understand tissue macrophage heterogeneity.


Assuntos
Receptor 2 de Folato/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Macrófagos/imunologia , Proteínas de Membrana/imunologia , Receptores CCR2/imunologia , Proteínas de Transporte Vesicular/imunologia , Animais , Estágios do Ciclo de Vida/imunologia , Ativação de Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores CCR2/deficiência
2.
Immunity ; 54(9): 2057-2071.e6, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34363749

RESUMO

Hypertension affects one-third of the world's population, leading to cardiac dysfunction that is modulated by resident and recruited immune cells. Cardiomyocyte growth and increased cardiac mass are essential to withstand hypertensive stress; however, whether immune cells are involved in this compensatory cardioprotective process is unclear. In normotensive animals, single-cell transcriptomics of fate-mapped self-renewing cardiac resident macrophages (RMs) revealed transcriptionally diverse cell states with a core repertoire of reparative gene programs, including high expression of insulin-like growth factor-1 (Igf1). Hypertension drove selective in situ proliferation and transcriptional activation of some cardiac RM states, directly correlating with increased cardiomyocyte growth. During hypertension, inducible ablation of RMs or selective deletion of RM-derived Igf1 prevented adaptive cardiomyocyte growth, and cardiac mass failed to increase, which led to cardiac dysfunction. Single-cell transcriptomics identified a conserved IGF1-expressing macrophage subpopulation in human cardiomyopathy. Here we defined the absolute requirement of RM-produced IGF-1 in cardiac adaptation to hypertension.


Assuntos
Adaptação Fisiológica/fisiologia , Hipertensão/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Macrófagos/metabolismo , Remodelação Ventricular/fisiologia , Animais , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Humanos , Hipertensão/complicações , Hipertensão/imunologia , Lactente , Masculino , Camundongos , Pessoa de Meia-Idade , Miocárdio/imunologia , Miocárdio/metabolismo , Miocárdio/patologia
3.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33372158

RESUMO

Macrophages are the principal immune cells of the epididymis and testis, but their origins, heterogeneity, development, and maintenance are not well understood. Here, we describe distinct populations of epididymal and testicular macrophages that display an organ-specific cellular identity. Combining in vivo fate-mapping, chimeric and parabiotic mouse models with in-depth cellular analyses, we found that CD64hiMHCIIlo and CD64loMHCIIhi macrophage populations of epididymis and testis arise sequentially from yolk sac erythro-myeloid progenitors, embryonic hematopoiesis, and nascent neonatal monocytes. While monocytes were the major developmental source of both epididymal and testicular macrophages, both populations self-maintain in the steady-state independent of bone marrow hematopoietic precursors. However, after radiation-induced macrophage ablation or during infection, bone marrow-derived circulating monocytes are recruited to the epididymis and testis, giving rise to inflammatory macrophages that promote tissue damage. These results define the layered ontogeny, maintenance and inflammatory response of macrophage populations in the male reproductive organs.


Assuntos
Infertilidade Masculina/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Animais , Diferenciação Celular , Linhagem da Célula , Epididimo/imunologia , Epididimo/metabolismo , Infertilidade Masculina/metabolismo , Infertilidade Masculina/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/imunologia , Testículo/imunologia , Testículo/metabolismo
4.
Nat Immunol ; 20(1): 29-39, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30538339

RESUMO

Macrophages promote both injury and repair after myocardial infarction, but discriminating functions within mixed populations remains challenging. Here we used fate mapping, parabiosis and single-cell transcriptomics to demonstrate that at steady state, TIMD4+LYVE1+MHC-IIloCCR2- resident cardiac macrophages self-renew with negligible blood monocyte input. Monocytes partially replaced resident TIMD4-LYVE1-MHC-IIhiCCR2- macrophages and fully replaced TIMD4-LYVE1-MHC-IIhiCCR2+ macrophages, revealing a hierarchy of monocyte contribution to functionally distinct macrophage subsets. Ischemic injury reduced TIMD4+ and TIMD4- resident macrophage abundance, whereas CCR2+ monocyte-derived macrophages adopted multiple cell fates within infarcted tissue, including those nearly indistinguishable from resident macrophages. Recruited macrophages did not express TIMD4, highlighting the ability of TIMD4 to track a subset of resident macrophages in the absence of fate mapping. Despite this similarity, inducible depletion of resident macrophages using a Cx3cr1-based system led to impaired cardiac function and promoted adverse remodeling primarily within the peri-infarct zone, revealing a nonredundant, cardioprotective role of resident cardiac macrophages.


Assuntos
Macrófagos/fisiologia , Infarto do Miocárdio/imunologia , Miocárdio/patologia , Animais , Receptor 1 de Quimiocina CX3C/metabolismo , Diferenciação Celular , Linhagem da Célula , Autorrenovação Celular , Perfilação da Expressão Gênica , Antígenos de Histocompatibilidade Classe II/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Parabiose , Receptores CCR2/genética , Receptores CCR2/metabolismo , Análise de Célula Única , Remodelação Ventricular , Proteínas de Transporte Vesicular/metabolismo
5.
Mol Cell Biochem ; 435(1-2): 163-173, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28526936

RESUMO

Emerging evidence suggests that long non-coding RNAs (lncRNAs) represent a cellular hub coordinating various cellular processes that are critical in health and disease. Mechanical stress triggers changes in vascular smooth muscle cells (VSMCs) that in turn contribute to pathophysiological changes within the vasculature. We sought to evaluate the role that lncRNAs play in mechanical stretch-induced alterations of human aortic smooth muscle cells (HASMCs). RNA (lncRNA and mRNA) samples isolated from HASMCs that had been subjected to 10 or 20% elongation (1 Hz) for 24 h were profiled with the Arraystar Human LncRNA Microarray V3.0. LncRNA expression was quantified in parallel via qRT-PCR. Of the 30,586 human lncRNAs screened, 580 were differentially expressed (DE, P < 0.05) in stretched HASMCs. Amongst the 26,109 protein-coding transcripts evaluated, 25 of those DE were associated with 25 of the aforementioned DE lncRNAs (P < 0.05). Subsequent Kyoto Encyclopedia of Genes and Genomes analysis revealed that the DE mRNAs were largely associated with the tumor necrosis factor signaling pathway and inflammation. Gene Ontology analysis indicated that the DE mRNAs were associated with cell differentiation, stress response, and response to external stimuli. We describe the first transcriptome profile of stretch-induced changes in HASMCs and provide novel insights into the regulatory switches that may be fundamental in governing aberrant VSMC remodeling.


Assuntos
Aorta/metabolismo , Perfilação da Expressão Gênica , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Longo não Codificante/biossíntese , Estresse Mecânico , Aorta/citologia , Humanos , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia
6.
Am J Physiol Lung Cell Mol Physiol ; 312(2): L208-L216, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27913427

RESUMO

Bronchopulmonary dysplasia (BPD) is a chronic lung injury characterized by impaired alveologenesis that may persist into adulthood. Rat models of BPD using varying degrees of hyperoxia to produce injury either cause early mortality or spontaneously recover following removal of the inciting stimulus, thus limiting clinical relevance. We sought to refine an established rat model induced by exposure to 60% O2 from birth by following hyperoxia with intermittent hypoxia (IH). Rats exposed from birth to air or 60% O2 until day 14 were recovered in air with or without IH (FIO2 = 0.10 for 10 min every 6 h) until day 28 Animals exposed to 60% O2 and recovered in air had no evidence of abnormal lung morphology on day 28 or at 10-12 wk. In contrast, 60% O2-exposed animals recovered in IH had persistently increased mean chord length, more dysmorphic septal crests, and fewer peripheral arteries. Recovery in IH also increased pulmonary vascular resistance, Fulton index, and arterial wall thickness. IH-mediated abnormalities in lung structure (but not pulmonary hypertension) persisted when reexamined at 10-12 wk, accompanied by increased pulmonary vascular reactivity and decreased exercise tolerance. Increased mean chord length secondary to IH was prevented by treatment with a peroxynitrite decomposition catalyst [5,10,15,20-Tetrakis(4-sulfonatophenyl)-21H,23H-porphyrin iron (III) chloride, 30 mg/kg/day, days 14-28], an effect accompanied by fewer inflammatory cells. We conclude that IH during recovery from hyperoxia-induced injury prevents recovery of alveologenesis and leads to changes in lung and pulmonary vascular function lasting into adulthood, thus more closely mimicking contemporary BPD.


Assuntos
Displasia Broncopulmonar/complicações , Displasia Broncopulmonar/patologia , Hiperóxia/complicações , Hipóxia/complicações , Lesão Pulmonar/complicações , Alvéolos Pulmonares/crescimento & desenvolvimento , Alvéolos Pulmonares/patologia , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Catálise , Modelos Animais de Doenças , Feminino , Hiperóxia/patologia , Hipertensão Pulmonar/complicações , Hipóxia/patologia , Lesão Pulmonar/patologia , Masculino , Metaloporfirinas/farmacologia , Ácido Peroxinitroso/metabolismo , Condicionamento Físico Animal , Pneumonia/complicações , Ratos Sprague-Dawley
7.
Am J Physiol Lung Cell Mol Physiol ; 311(2): L292-302, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27317685

RESUMO

Systemically-administered bleomycin causes inflammation, arrested lung growth, and pulmonary hypertension (PHT) in the neonatal rat, similar to human infants with severe bronchopulmonary dysplasia (BPD). Leukotrienes (LTs) are inflammatory lipid mediators produced by multiple cell types in the lung. The major LTs, LTB4 and cysteinyl LTs, are suggested to contribute to BPD, but their specific roles remain largely unexplored in experimental models. We hypothesized that LTs are increased in bleomycin-induced BPD-like injury, and that inhibition of LT production would prevent inflammatory cell influx and thereby ameliorate lung injury. Rat pups were exposed to bleomycin (1 mg·kg(-1)·day(-1) ip) or vehicle (control) from postnatal days 1-14 and were treated with either zileuton (5-lipoxygenase inhibitor), montelukast (cysteinyl LT1 receptor antagonist), or SC57461A (LTA4 hydrolase inhibitor) 10 mg·kg(-1)·day(-1) ip. Bleomycin led to increased lung content of LTB4, but not cysteinyl LTs. Bleomycin-induced increases in tissue neutrophils and macrophages and lung contents of LTB4 and tumor necrosis factor-α were all prevented by treatment with zileuton. Treatment with zileuton or SC57461A also prevented the hemodynamic and structural markers of chronic PHT, including raised pulmonary vascular resistance, increased Fulton index, and arterial wall remodeling. However, neither treatment prevented impaired alveolarization or vascular hypoplasia secondary to bleomycin. Treatment with montelukast had no effect on macrophage influx, PHT, or on abnormal lung structure. We conclude that LTB4 plays a crucial role in lung inflammation and PHT in experimental BPD. Agents targeting LTB4 or LTB4-mediated signaling may have utility in infants at risk of developing BPD-associated PHT.


Assuntos
Displasia Broncopulmonar/imunologia , Hipertensão Pulmonar/imunologia , Leucotrieno B4/fisiologia , Macrófagos/imunologia , Animais , Animais Recém-Nascidos , Bleomicina , Displasia Broncopulmonar/induzido quimicamente , Displasia Broncopulmonar/metabolismo , Movimento Celular/imunologia , Feminino , Expressão Gênica , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/metabolismo , Ratos Sprague-Dawley
8.
Am J Physiol Lung Cell Mol Physiol ; 309(1): L53-62, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26136527

RESUMO

Neutrophil (PMNL) influx precedes lung macrophage (LM) influx into the lung following exposure of newborn pups to 60% O2. We hypothesized that PMNL were responsible for the signals leading to LM influx. This was confirmed when inhibition of PMNL influx with a CXC chemokine receptor-2 antagonist, SB-265610, also prevented the 60% O2-dependent LM influx, LM-derived nitrotyrosine formation, and pruning of small arterioles. Exposure to 60% O2 was associated with increased lung contents of neutrophil elastase and α-elastin, a marker of denatured elastin, and a decrease in elastin fiber density. This led us to speculate that neutrophil elastase-induced elastin fragments were the chemokines that led to a LM influx into the 60% O2-exposed lung. Inhibition of neutrophil elastase with sivelestat or elafin attenuated the LM influx. Sivelestat also attenuated the 60% O2-induced decrease in elastin fiber density. Daily injections of pups with an antibody to α-elastin prevented the 60% O2-dependent LM influx, impaired alveologenesis, and impaired small vessel formation. This suggests that neutrophil elastase inhibitors may protect against neonatal lung injury not only by preventing structural elastin degradation, but also by blocking elastin fragment-induced LM influx, thus preventing tissue injury from LM-derived peroxynitrite formation.


Assuntos
Elastina/metabolismo , Elastase de Leucócito/metabolismo , Macrófagos/imunologia , Neutrófilos/imunologia , Oxigênio/toxicidade , Animais , Animais Recém-Nascidos , Movimento Celular/imunologia , Elafina/farmacologia , Elastina/imunologia , Feminino , Glicina/análogos & derivados , Glicina/farmacologia , Elastase de Leucócito/antagonistas & inibidores , Pulmão/patologia , Lesão Pulmonar/imunologia , Exposição Materna , Oxigênio/farmacologia , Ácido Peroxinitroso/biossíntese , Compostos de Fenilureia/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Interleucina-8B/antagonistas & inibidores , Sulfonamidas/farmacologia , Triazóis/farmacologia , Remodelação Vascular
9.
Am J Physiol Lung Cell Mol Physiol ; 308(6): L503-10, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25595650

RESUMO

Arginase is an enzyme that limits substrate L-arginine bioavailability for the production of nitric oxide by the nitric oxide synthases and produces L-ornithine, which is a precursor for collagen formation and tissue remodeling. We studied the pulmonary vascular effects of arginase inhibition in an established model of repeated systemic bleomycin sulfate administration in neonatal rats that results in pulmonary hypertension and lung injury mimicking the characteristics typical of bronchopulmonary dysplasia. We report that arginase expression is increased in the lungs of bleomycin-exposed neonatal rats and that treatment with the arginase inhibitor amino-2-borono-6-hexanoic acid prevented the bleomycin-induced development of pulmonary hypertension and deposition of collagen. Arginase inhibition resulted in increased L-arginine and L-arginine bioavailability and increased pulmonary nitric oxide production. Arginase inhibition also normalized the expression of inducible nitric oxide synthase, and reduced bleomycin-induced nitrative stress while having no effect on bleomycin-induced inflammation. Our data suggest that arginase is a promising target for therapeutic interventions in neonates aimed at preventing lung vascular remodeling and pulmonary hypertension.


Assuntos
Aminocaproatos/farmacologia , Antibióticos Antineoplásicos/efeitos adversos , Arginase/antagonistas & inibidores , Bleomicina/efeitos adversos , Compostos de Boro/farmacologia , Colágeno/metabolismo , Hipertensão Pulmonar , Pulmão/enzimologia , Remodelação Vascular/efeitos dos fármacos , Animais , Antibióticos Antineoplásicos/farmacologia , Arginase/metabolismo , Arginina/metabolismo , Bleomicina/farmacologia , Displasia Broncopulmonar/induzido quimicamente , Displasia Broncopulmonar/enzimologia , Displasia Broncopulmonar/patologia , Displasia Broncopulmonar/prevenção & controle , Modelos Animais de Doenças , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/enzimologia , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/prevenção & controle , Pulmão/patologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/enzimologia , Lesão Pulmonar/patologia , Lesão Pulmonar/prevenção & controle , Óxido Nítrico/metabolismo , Ratos , Ratos Sprague-Dawley
10.
Free Radic Biol Med ; 69: 35-49, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24423485

RESUMO

Chronic pulmonary hypertension in the neonate and infant frequently presents with right-ventricular (RV) failure. Current clinical management may include protracted treatment with inhaled nitric oxide (iNO), with the goal of reducing RV afterload. We have previously reported that prolonged exposure to iNO causes RV systolic dysfunction in the chronic hypoxia-exposed juvenile rat, which was prevented by a peroxynitrite decomposition catalyst. Given that inhalation of CO2 (therapeutic hypercapnia) may limit oxidative stress and upregulated cytokine expression in the lung and other organs, we hypothesized that therapeutic hypercapnia would attenuate cytokine-mediated nitric oxide synthase (NOS) upregulation, thus limiting peroxynitrite generation. Sprague-Dawley rat pups were exposed to chronic hypoxia (13% O2) from postnatal day 1 to 21, while receiving iNO (20 ppm) from day 14 to 21, with or without therapeutic hypercapnia (10% CO2). Therapeutic hypercapnia completely normalized RV systolic function, RV hypertrophy, and remodeling of pulmonary resistance arteries in animals exposed to iNO. Inhaled nitric oxide-mediated increases in RV peroxynitrite, apoptosis, and contents of tumor necrosis factor (TNF)-α, interleukin (IL)-1α, and NOS-2 were all attenuated by therapeutic hypercapnia. Inhibition of NOS-2 activity with 1400 W (1 mg/kg/day) prevented iNO-mediated upregulation of peroxynitrite and led to improved RV systolic function. Blockade of IL-1 receptor signaling with anakinra (500 mg/kg/day) decreased NOS-2 content and had similar effects compared to NOS-2 inhibition on iNO-mediated effects, whereas blockade of TNF-α signaling with etanercept (0.4 mg/kg on alternate days) had no effects on these parameters. We conclude that therapeutic hypercapnia prevents the adverse effects of sustained exposure to iNO on RV systolic function by limiting IL-1-mediated NOS-2 upregulation and consequent nitration. Therapeutic hypercapnia also acts synergistically with iNO in normalizing RV hypertrophy, vascular remodeling, and raised pulmonary vascular resistance secondary to chronic hypoxia.


Assuntos
Dióxido de Carbono/sangue , Hipercapnia/sangue , Hipertensão Pulmonar/terapia , Hipertrofia Ventricular Direita/terapia , Animais , Humanos , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/metabolismo , Hipertrofia Ventricular Direita/induzido quimicamente , Hipertrofia Ventricular Direita/patologia , Interleucina-1/metabolismo , Óxido Nítrico/toxicidade , Óxido Nítrico Sintase/metabolismo , Ratos , Fator de Necrose Tumoral alfa/metabolismo
11.
Am J Respir Cell Mol Biol ; 50(1): 61-73, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23947621

RESUMO

Bleomycin-induced lung injury is characterized in the neonatal rat by inflammation dominated by neutrophils and macrophages, inhibited distal airway and vascular development, and pulmonary hypertension, similar to human infants with severe bronchopulmonary dysplasia. Rho-kinase (ROCK) is known to mediate lung injury in adult animals via stimulatory effects on inflammation. We therefore hypothesized that inhibition of ROCK may ameliorate bleomycin-induced lung injury in the neonatal rat. Pups received daily intraperitoneal bleomycin or saline from Postnatal Days 1 through 14 with or without Y-27632, a ROCK inhibitor. Treatment with Y-27632 prevented bleomycin-induced pulmonary hypertension, as evidenced by normalized pulmonary vascular resistance, decreased right-ventricular hypertrophy, and attenuated remodeling of pulmonary resistance arteries. Bleomycin-induced changes in distal lung architecture, including septal thinning, inhibited alveolarization, and decreased numbers of peripheral arteries and capillaries, were partially or completely normalized by Y-27632. Treatment with Y-27632 or a CXCR2 antagonist, SB265610, also abrogated tissue neutrophil influx, while having no effect on macrophages. However, treatment with SB265610 did not prevent bleomycin-induced lung injury. Lung content of angiostatic thrombospondin-1 (TSP1) was increased significantly in the lungs of bleomycin-exposed animals, and was completely attenuated by treatment with Y-27632. Thrombin-stimulated TSP1 production by primary cultured rat pulmonary artery endothelial cells was also attenuated by Y-27632. Taken together, our findings suggest a preventive effect of Y-27632 on bleomycin-mediated injury by a mechanism unrelated to inflammatory cells. Our data suggest that improvements in lung morphology may have been related to indirect stimulatory effects on angiogenesis via down-regulation of TSP1.


Assuntos
Inibidores Enzimáticos/farmacologia , Lesão Pulmonar/prevenção & controle , Pneumonia/diagnóstico por imagem , Pneumonia/patologia , Quinases Associadas a rho/antagonistas & inibidores , Amidas/farmacologia , Animais , Animais Recém-Nascidos , Bleomicina/efeitos adversos , Quimiocinas/metabolismo , Regulação para Baixo/efeitos dos fármacos , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/prevenção & controle , Hipertrofia Ventricular Direita/tratamento farmacológico , Hipertrofia Ventricular Direita/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/metabolismo , Macrófagos/diagnóstico por imagem , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Neutrófilos/patologia , Pneumonia/metabolismo , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , Artéria Pulmonar/patologia , Piridinas/farmacologia , Radiografia , Ratos , Ratos Sprague-Dawley , Trombospondina 1/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Resistência Vascular/efeitos dos fármacos , Quinases Associadas a rho/metabolismo
12.
Pediatr Pulmonol ; 49(10): 991-1002, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24273102

RESUMO

RATIONALE: Use of the anti-inflammatory agent dexamethasone in premature infants with bronchopulmonary dysplasia has been curtailed, and no alternative anti-inflammatory agents are approved for this use. Our objective was to use a neonatal rat model of bronchopulmonary dysplasia to determine if an highly selective cyclooxygenase-2 inhibitor, 5,5-dimethyl-3-(3-fluorophenyl)4-(4-methylsulfonyl)phenyl-2(5H)-furanone (DFU; 10 µg/g body weight), could prevent inflammatory cell influx and protect against lung injury. METHODS: Neonatal rats exposed to air or 60% O2 for 14 days from birth either received daily i.p. injections of (i) vehicle or DFU or (ii) vehicle or an EP(1) receptor antagonist, SC-19220. RESULTS: DFU attenuated the lung macrophage and neutrophil influx, prevented interstitial thickening and prevented the loss of peripheral blood vessels induced by 60% O2 , but did not protect against the variance in alveolar diameter induced by 60% O2 . Exposure to 60% O2 caused both an increase in lung prostaglandin E2 content and a reduction in lung mesenchymal cell mass which was reversed by DFU. Prostaglandin E2 binding to the EP(1) receptor inhibited DNA synthesis in cultures of lung fibroblasts in a dose dependent fashion. Treatment with SC-19220 attenuated the reduction in lung mesenchymal mass observed following exposure of rat pups to 60% O2 . CONCLUSIONS: An highly selective cyclooxygenase-2 inhibitor is an effective anti-inflammatory substitute for dexamethasone for preventing phagocyte influx into the neonatal lung during 60% O2 -mediated lung injury, and can modify the severity of that injury.


Assuntos
Displasia Broncopulmonar/tratamento farmacológico , Inibidores de Ciclo-Oxigenase 2/farmacologia , Furanos/farmacologia , Oxigênio/administração & dosagem , Animais , Animais Recém-Nascidos , Dinoprostona/metabolismo , Pulmão/metabolismo , Ratos Sprague-Dawley
13.
Am J Physiol Lung Cell Mol Physiol ; 303(1): L75-87, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22582114

RESUMO

Bleomycin-induced lung injury is characterized in the neonatal rat by inflammation, arrested lung growth, and pulmonary hypertension (PHT), as observed in human infants with severe bronchopulmonary dysplasia. Inhalation of CO(2) (therapeutic hypercapnia) has been described to limit cytokine production and to have anti-inflammatory effects on the injured lung; we therefore hypothesized that therapeutic hypercapnia would prevent bleomycin-induced lung injury. Spontaneously breathing rat pups were treated with bleomycin (1 mg/kg/d ip) or saline vehicle from postnatal days 1-14 while being continuously exposed to 5% CO(2) (Pa(CO(2)) elevated by 15-20 mmHg), 7% CO(2) (Pa(CO(2)) elevated by 35 mmHg), or normocapnia. Bleomycin-treated animals exposed to 7%, but not 5%, CO(2), had significantly attenuated lung tissue macrophage influx and PHT, as evidenced by normalized pulmonary vascular resistance and right ventricular systolic function, decreased right ventricular hypertrophy, and attenuated remodeling of pulmonary resistance arteries. The level of CO(2) neither prevented increased tissue neutrophil influx nor led to improvements in decreased lung weight, septal thinning, impaired alveolarization, or decreased numbers of peripheral arteries. Bleomycin led to increased expression and content of lung tumor necrosis factor (TNF)-α, which was found to colocalize with tissue macrophages and to be attenuated by exposure to 7% CO(2). Inhibition of TNF-α signaling with the soluble TNF-2 receptor etanercept (0.4 mg/kg ip from days 1-14 on alternate days) prevented bleomycin-induced PHT without decreasing tissue macrophages and, similar to CO(2), had no effect on arrested alveolar development. Our findings are consistent with a preventive effect of therapeutic hypercapnia with 7% CO(2) on bleomycin-induced PHT via attenuation of macrophage-derived TNF-α. Neither tissue macrophages nor TNF-α appeared to contribute to arrested lung development induced by bleomycin. That 7% CO(2) normalized pulmonary vascular resistance and right ventricular function without improving inhibited airway and vascular development suggests that vascular hypoplasia does not contribute significantly to functional changes of PHT in this model.


Assuntos
Hipercapnia/fisiopatologia , Hipertensão Pulmonar/prevenção & controle , Macrófagos/metabolismo , Alvéolos Pulmonares/fisiopatologia , Artéria Pulmonar/fisiopatologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo , Animais , Animais Recém-Nascidos/metabolismo , Animais Recém-Nascidos/fisiologia , Bleomicina/toxicidade , Dióxido de Carbono/administração & dosagem , Dióxido de Carbono/sangue , Hipercapnia/sangue , Hipercapnia/induzido quimicamente , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/metabolismo , Hipertrofia Ventricular Direita/metabolismo , Hipertrofia Ventricular Direita/fisiopatologia , Inflamação/metabolismo , Inflamação/fisiopatologia , Pulmão/metabolismo , Pulmão/fisiopatologia , Lesão Pulmonar/metabolismo , Lesão Pulmonar/fisiopatologia , Lesão Pulmonar/prevenção & controle , Neutrófilos/metabolismo , Alvéolos Pulmonares/metabolismo , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , Ratos , Função Ventricular Direita/efeitos dos fármacos
14.
Am J Physiol Lung Cell Mol Physiol ; 300(3): L319-29, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21148793

RESUMO

During early postnatal alveolar formation, the lung tissue of rat pups undergoes a physiological remodeling involving apoptosis of distal lung cells. Exposure of neonatal rats to severe hyperoxia (≥95% O(2)) both arrests lung growth and results in increased lung cell apoptosis. In contrast, exposure to moderate hyperoxia (60% O(2)) for 14 days does not completely arrest lung cell proliferation and is associated with parenchymal thickening. On the basis of similarities in lung architecture observed following either exposure to 60% O(2), or pharmacological inhibition of physiological apoptosis, we hypothesized that exposure to 60% O(2) would result in an inhibition of physiological lung cell apoptosis. Consistent with this hypothesis, we observed that the parenchymal thickening induced by exposure to 60% O(2) was associated with decreased numbers of apoptotic cells, increased expressions of the antiapoptotic regulator Bcl-xL, and the putative antiapoptotic protein survivin, and decreased expressions of the proapoptotic cleaved caspases-3 and -7. In summary, exposure of the neonatal rat lung to moderate hyperoxia results in an inhibition of physiological apoptosis, which contributes to the parenchymal thickening observed in the resultant lung injury.


Assuntos
Apoptose/efeitos dos fármacos , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/patologia , Oxigênio/farmacologia , Transdução de Sinais/efeitos dos fármacos , Ar , Animais , Animais Recém-Nascidos , Western Blotting , Caspase 3/metabolismo , Contagem de Células , Morte Celular/efeitos dos fármacos , Feminino , Imuno-Histoquímica , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Pulmão/patologia , Ratos , Ratos Sprague-Dawley , Estaurosporina/farmacologia
15.
Free Radic Res ; 45(3): 359-65, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21034359

RESUMO

Neonatal lung injury has been induced experimentally by infusion of multivitamin-containing light-exposed parenteral nutrition (PN) solutions. The objective was to explore the role of ascorbate in toxic effects of light-exposed PN on primary cultured foetal rat lung epithelial cells. Hydroperoxides were measured in 3% amino acid solutions at baseline, immediately after addition of either multivitamins or ascorbate alone (400 µg/mL) and again after a 24-h period of exposure to (or protection from) ambient light. Cellular toxicity was assessed by [C(14)]adenine release. Multivitamins or ascorbate alone increased hydroperoxides in PN, which was attenuated by light protection. Light-exposed PN containing multivitamins was more toxic to cells than baseline or light-protected PN. Exposure to ascorbate at concentrations both lower (< 5 µg/mL) and higher (> 1000 µg/mL) than normally contained in PN-induced oxidant-mediated cell death, as indicated by protective effects of hydroperoxide and hydroxyl radical scavengers. This study concludes that ascorbate generates toxic amounts of peroxide in PN solutions. The types and physiological importance of hydroperoxides induced by pro-oxidant effects of ascorbate require further evaluation in vivo.


Assuntos
Ácido Ascórbico/toxicidade , Células Epiteliais/efeitos dos fármacos , Pulmão/citologia , Soluções de Nutrição Parenteral/toxicidade , Animais , Células Cultivadas , Sequestradores de Radicais Livres/farmacologia , Peróxido de Hidrogênio/farmacologia , Luz , Ratos , Ratos Sprague-Dawley
16.
Free Radic Biol Med ; 49(7): 1182-91, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20647043

RESUMO

Exposure of newborn rats to 60% O2 for 14days results in a chronic neonatal lung injury characterized by parenchymal thickening, impaired alveolarization, evidence of pulmonary hypertension, and pulmonary vascular pruning. The contribution of peroxynitrite to this injury was assessed by treating pups with a peroxynitrite decomposition catalyst, 5,10,15,20-tetrakis(4-sulfonatophenyl)porphyrinato iron(III) chloride (FeTPPS), at 30microg/g/day. Body and lung weights and postfixation lung volumes were all slightly, but significantly, reduced by exposure to 60% O2 and this was attenuated by a concurrent FeTPPS intervention. The FeTPPS intervention had no impact on increased neutrophil or macrophage influx into the lung, but attenuated 60% O2-induced reductions in the lung contents of vascular endothelial-derived growth factor, its receptor-2, and angiopoietin and increases in 8-isoprostane and preproendothelin-1. The 60% O2-induced parenchymal thickening and impairment of alveologenesis, as well as vascular pruning and peripheral vessel medial wall thickening, were attenuated by FeTPPS, despite a persistent inflammatory cell influx. Pups exposed to 60% O2 and treated with FeTPPS had enhanced alveolar formation relative to control pups. We conclude that peroxynitrite plays a critical role in the development of chronic neonatal lung injury.


Assuntos
Lesão Pulmonar/prevenção & controle , Pulmão/metabolismo , Metaloporfirinas/administração & dosagem , Ácido Peroxinitroso/metabolismo , Angiopoietina-1/genética , Angiopoietina-1/metabolismo , Animais , Animais Recém-Nascidos , Catálise , Hipertensão Pulmonar , Imuno-Histoquímica , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Pulmão/patologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/fisiopatologia , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Metaloporfirinas/química , Neovascularização Fisiológica/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Neutrófilos/patologia , Oxigênio/administração & dosagem , Ácido Peroxinitroso/química , Ratos , Ratos Sprague-Dawley , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
17.
Pediatr Res ; 67(2): 177-82, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19858775

RESUMO

Rho-kinase (ROCK) inhibitors prevent pulmonary hypertension (PHT) in adult rodents, but little is known about their effects on the neonatal lung. Our objective was to examine the effects of ROCK inhibition on chronic hypoxia (CH)-induced PHT and abnormal lung structure in the neonatal rat. Pups were exposed to air or CH from postnatal d 1-14 while receiving Y-27632 (5 or 10 mg x kg(-1) x d(-1)), fasudil (20 mg x kg(-1) x d(-1)), or saline intraperitoneally. Relative to air, CH-exposed pups had increased pulmonary vascular resistance, right ventricular hypertrophy, arterial medial wall thickening, and abnormal distal airway morphology characterized by septal thinning and decreased secondary septation. Treatment with 10 mg/kg Y-27632 or fasudil attenuated the structural and hemodynamic changes of PHT while having no effect on septal thinning or inhibited secondary septation. In addition, Y-27632 (10 mg/kg) and fasudil augmented CH-induced somatic growth restriction. Pulmonary arteries of CH-exposed pups had increased ROCK activity, up-regulated expression of PDGF-BB and increased smooth muscle DNA synthesis, all of which were attenuated by treatment with 10 mg/kg Y-27632. Systemically administered ROCK inhibitors prevented PHT in the CH-exposed neonatal rat but at the cost of inhibited somatic growth. Limiting effects on vascular remodeling likely resulted, in major part, from attenuated vascular PDGF-BB/beta-receptor signaling.


Assuntos
1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , Amidas/farmacologia , Anti-Hipertensivos/farmacologia , Hiperóxia/tratamento farmacológico , Hipertensão Pulmonar/prevenção & controle , Pulmão/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Quinases Associadas a rho/antagonistas & inibidores , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Remodelação das Vias Aéreas/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Becaplermina , Peso Corporal , Doença Crônica , Replicação do DNA , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Hiperóxia/complicações , Hiperóxia/enzimologia , Hiperóxia/patologia , Hiperóxia/fisiopatologia , Hipertensão Pulmonar/enzimologia , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/fisiopatologia , Hipertrofia Ventricular Direita/enzimologia , Hipertrofia Ventricular Direita/etiologia , Hipertrofia Ventricular Direita/prevenção & controle , Pulmão/irrigação sanguínea , Pulmão/enzimologia , Pulmão/patologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/patologia , Tamanho do Órgão , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-sis , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/enzimologia , Artéria Pulmonar/patologia , Ratos , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Resistência Vascular/efeitos dos fármacos , Quinases Associadas a rho/metabolismo
18.
Am J Physiol Lung Cell Mol Physiol ; 297(5): L920-30, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19749000

RESUMO

Permissive hypercapnia, achieved using low tidal volume ventilation, has been an effective protective strategy in patients with acute respiratory distress syndrome. To date, no such protective effect has been demonstrated for the chronic neonatal lung injury, bronchopulmonary dysplasia. The objective of our study was to determine whether evolving chronic neonatal lung injury, using a rat model, is resistant to the beneficial effects of hypercapnia or simply requires a less conservative approach to hypercapnia than that applied clinically to date. Neonatal rats inhaled air or 60% O2 for 14 days with or without 5.5% CO2. Lung parenchymal neutrophil and macrophage numbers were significantly increased by hyperoxia alone, which was associated with interstitial thickening and reduced secondary crest formation. The phagocyte influx, interstitial thickening, and impaired alveolar formation were significantly attenuated by concurrent hypercapnia. Hyperoxic pups that received 5.5% CO2 had a significant increase in alveolar number relative to air-exposed pups. Increased tyrosine nitration, a footprint for peroxynitrite-mediated reactions, arteriolar medial wall thickening, and both reduced small peripheral pulmonary vessel number and VEGF and angiopoietin-1 (Ang-1) expression, which were observed with hyperoxia, was attenuated by concurrent hypercapnia. We conclude that evolving chronic neonatal lung injury in a rat model is responsive to the beneficial effects of hypercapnia. Inhaled 5.5% CO2 provided a significant degree of protection against parenchymal and vascular injury in an animal model of chronic neonatal lung injury likely due, at least in part, to its inhibition of a phagocyte influx.


Assuntos
Vasos Sanguíneos/fisiologia , Hipercapnia/complicações , Hipercapnia/fisiopatologia , Lesão Pulmonar/fisiopatologia , Lesão Pulmonar/terapia , Actinas/metabolismo , Ar , Animais , Animais Recém-Nascidos , Gasometria , Vasos Sanguíneos/efeitos dos fármacos , Dióxido de Carbono/farmacologia , Elastina/metabolismo , Imunofluorescência , Frequência Cardíaca/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Pulmão/patologia , Lesão Pulmonar/complicações , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/enzimologia , Tamanho do Órgão/efeitos dos fármacos , Peroxidase/metabolismo , Ratos , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo , Fator de von Willebrand/metabolismo
19.
Pediatr Res ; 66(3): 260-5, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19542903

RESUMO

IL-1 beta, a proinflammatory cytokine, may contribute to the development of the chronic neonatal lung injury, bronchopulmonary dysplasia. Chronic neonatal lung injury was induced in rats, by exposure to 60% O2 for 14 d from birth, to determine whether pulmonary IL-1 expression was up-regulated and, if so, whether a daily s.c. IL-1 receptor antagonist injections would be protective. Exposure to 60% O2 for 14 d caused pulmonary neutrophil and macrophage influx, increased tissue fraction and tyrosine nitration, reduced VEGF-A and angiopoietin-1 expression, and reduced small vessel (20-65 microm) and alveolar numbers. Lung IL-1 alpha and -1 beta contents were increased after a 4-d exposure to 60% O2. IL-1 receptor antagonist treatment attenuated the 60% O2-dependent neutrophil influx, the increased tissue fraction, and the reduced alveolar number. Treatment did not restore VEGF-A or angiopoietin-1 expression and only partially attenuated the reduced vessel number in 60% O2-exposed pups. It also caused a paradoxical increase in macrophage influx and a reduction in small vessels in air-exposed pups. We conclude that antagonism of IL-1-mediated effects can, in major part, protect against lung injury in a rat model of 60% O2-induced chronic neonatal lung injury.


Assuntos
Lesão Pulmonar/etiologia , Oxigênio/efeitos adversos , Receptores de Interleucina-1/metabolismo , Angiopoietina-1/metabolismo , Animais , Animais Recém-Nascidos , Peso Corporal , Proteína Antagonista do Receptor de Interleucina 1/metabolismo , Interleucina-1beta/metabolismo , Pulmão/citologia , Pulmão/metabolismo , Pulmão/patologia , Macrófagos/citologia , Macrófagos/metabolismo , Neutrófilos/citologia , Neutrófilos/metabolismo , Tamanho do Órgão , Fagócitos/metabolismo , Ratos , Tirosina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
20.
Am J Physiol Lung Cell Mol Physiol ; 294(2): L205-13, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18032699

RESUMO

Pulmonary hypertension (PHT) in neonates is often refractory to the current best therapy, inhaled nitric oxide (NO). The utility of a new class of pulmonary vasodilators, Rho-kinase (ROCK) inhibitors, has not been examined in neonatal animals. Our objective was to examine the activity and expression of RhoA/ROCK in normal and injured pulmonary arteries and to determine the short-term pulmonary hemodynamic (assessed by pulse wave Doppler) effects of ROCK inhibitors (15 mg/kg ip Y-27632 or 30 mg/kg ip fasudil) in two neonatal rat models of chronic PHT with pulmonary vascular remodeling (chronic hypoxia, 0.13 Fi(O(2)), or 1 mg.kg(-1).day(-1) ip chronic bleomycin for 14 days from birth). Activity of the RhoA/ROCK pathway and ROCK expression were increased in hypoxia- and bleomycin-induced PHT. In both models, severe PHT [characterized by raised pulmonary vascular resistance (PVR) and impaired right ventricular (RV) performance] did not respond acutely to inhaled NO (20 ppm for 15 min) or to a single bolus of a NO donor, 3-morpholinosydnonimine hydrochloride (SIN-1; 2 mug/kg ip). In contrast, a single intraperitoneal bolus of either ROCK inhibitor (Y-27632 or fasudil) completely normalized PVR but had no acute effect on RV performance. ROCK-mediated vasoconstriction appears to play a key role in chronic PHT in our two neonatal rat models. Inhibitors of ROCK have potential as a testable therapy in neonates with PHT that is refractory to NO.


Assuntos
Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/enzimologia , Óxido Nítrico/uso terapêutico , Vasodilatadores/uso terapêutico , Quinases Associadas a rho/antagonistas & inibidores , Administração por Inalação , Amidas/farmacologia , Animais , Animais Recém-Nascidos , Bleomicina , Movimento Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Hipertensão Pulmonar/fisiopatologia , Hipóxia , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Pulmão/patologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Óxido Nítrico/administração & dosagem , Óxido Nítrico/farmacologia , Doadores de Óxido Nítrico/farmacologia , Piridinas/farmacologia , Ratos , Ratos Sprague-Dawley , Resistência Vascular/efeitos dos fármacos , Função Ventricular Direita/efeitos dos fármacos , Proteína rhoA de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA