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

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Pediatr Res ; 85(5): 733, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30710117

RESUMO

Following publication of this article the authors noticed that affiliations were incorrectly assigned. The original article has now been updated so that the author "Joyce R. Javier" is associated with the "Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Department of Pediatrics, Division of General Pediatrics, Los Angeles, CA, USA", and the author "Vivek Balasubramaniam" is associated with the "Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, USA". This has been corrected in both the PDF and HTML versions of the article.

2.
Biochem Biophys Res Commun ; 503(4): 2653-2658, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30093115

RESUMO

Mesenchymal stromal cell (MSC) derived exosomes mediate tissue protection and regeneration in many injuries and diseases by modulating cell protein production, protecting from apoptosis, inhibiting inflammation, and increasing angiogenesis. In the present study, daily intraperitoneal injection of MSC-derived exosomes protected alveolarization and angiogenesis in a newborn rat model of bronchopulmonary dysplasia (BPD) induced by 14 days of neonatal hyperoxia exposure (85% O2). Exosome treatment during hyperoxia prevented disruption of alveolar growth, increased small blood vessel number, and inhibited right heart hypertrophy at P14, P21, and P56. In vitro, exosomes significantly increased tube-like network formation by HUVEC, in part through a VEGF mediated mechanism. In summary, daily intraperitoneal injection of exosomes increased blood vessel number and size in the lung through pro-angiogenic mechanisms. MSC-derived exosomes therefore have both anti-inflammatory and pro-angiogenic mechanism to protect the lung from hyperoxia induced lung and heart disease associated with BPD.


Assuntos
Displasia Broncopulmonar/prevenção & controle , Cardiomegalia/prevenção & controle , Exossomos/fisiologia , Hiperóxia/prevenção & controle , Células-Tronco Mesenquimais/química , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Animais Recém-Nascidos , Células da Medula Óssea/química , Células da Medula Óssea/citologia , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patologia , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Modelos Animais de Doenças , Exossomos/transplante , Feminino , Regulação da Expressão Gênica , Hiperóxia/genética , Hiperóxia/metabolismo , Hiperóxia/patologia , Injeções Intraperitoneais , Pulmão/irrigação sanguínea , Pulmão/metabolismo , Pulmão/patologia , Células-Tronco Mesenquimais/citologia , Neovascularização Fisiológica/genética , Oxigênio/toxicidade , Gravidez , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Fator A de Crescimento do Endotélio Vascular/agonistas , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
Am J Physiol Lung Cell Mol Physiol ; 306(7): L661-71, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24508729

RESUMO

Angiogenic expansion of the vasa vasorum (VV) is an important contributor to pulmonary vascular remodeling in the pathogenesis of pulmonary hypertension (PH). High proliferative potential endothelial progenitor-like cells have been described in vascular remodeling and angiogenesis in both systemic and pulmonary circulations. However, their role in hypoxia-induced pulmonary artery (PA) VV expansion in PH is not known. We hypothesized that profound PA VV neovascularization observed in a neonatal calf model of hypoxia-induced PH is due to increased numbers of subsets of high proliferative cells within the PA adventitial VV endothelial cells (VVEC). Using a single cell clonogenic assay, we found that high proliferative potential colony-forming cells (HPP-CFC) comprise a markedly higher percentage in VVEC populations isolated from the PA of hypoxic (VVEC-Hx) compared with control (VVEC-Co) calves. VVEC-Hx populations that comprised higher numbers of HPP-CFC also demonstrated markedly higher expression levels of CD31, CD105, and c-kit than VVEC-Co. In addition, significantly higher expression of CD31, CD105, and c-kit was observed in HPP-CFC vs. the VVEC of the control but not of hypoxic animals. HPP-CFC exhibited migratory and tube formation capabilities, two important attributes of angiogenic phenotype. Furthermore, HPP-CFC-Co and some HPP-CFC-Hx exhibited elevated telomerase activity, consistent with their high replicative potential, whereas a number of HPP-CFC-Hx exhibited impaired telomerase activity, suggestive of their senescence state. In conclusion, our data suggest that hypoxia-induced VV expansion involves an emergence of HPP-CFC populations of a distinct phenotype with increased angiogenic capabilities. These cells may serve as a potential target for regulating VVEC neovascularization.


Assuntos
Hipertensão Pulmonar/etiologia , Hipóxia/fisiopatologia , Neovascularização Patológica/patologia , Artéria Pulmonar/patologia , Vasa Vasorum/fisiopatologia , Animais , Animais Recém-Nascidos , Antígenos CD/metabolismo , Bovinos , Ensaios de Migração Celular , Proliferação de Células , Ensaio de Unidades Formadoras de Colônias , Hipertensão Pulmonar/fisiopatologia , Hipóxia/metabolismo , Masculino , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Telomerase/metabolismo
7.
J Biol Chem ; 287(38): 31994-2005, 2012 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-22854956

RESUMO

Cystathionine (R-S-(2-amino-2-carboxyethyl)-l-homocysteine) is a non-proteinogenic thioether containing amino acid. In mammals, cystathionine is formed as an intermediate of the transsulfuration pathway by the condensation of serine and homocysteine (Hcy) in a reaction catalyzed by cystathionine ß-synthase (CBS). Cystathionine is subsequently converted to cysteine plus ammonia and α-ketobutyrate by the action of cystathionine γ-lyase (CGL). Pathogenic mutations in CBS result in CBS-deficient homocystinuria (HCU) which, if untreated, results in mental retardation, thromboembolic complications and connective tissue disorders. Currently there is no known function for cystathionine other than serving as an intermediate in transsulfuration and to date, the possible contribution of the abolition of cystathionine synthesis to pathogenesis in HCU has not been investigated. Using both mouse and cell-culture models, we have found that cystathionine is capable of blocking the induction of hepatic steatosis and kidney injury, acute tubular necrosis, and apoptotic cell death by the endoplasmic reticulum stress inducing agent tunicamycin. Northern and Western blotting analysis indicate that the protective effects of cystathionine occur without any obvious alteration of the induction of the unfolded protein response. Our data constitute the first experimental evidence that the abolition of cystathionine synthesis may contribute to the pathology of HCU and that this compound has therapeutic potential for disease states where ER stress is implicated as a primary initiating pathogenic factor.


Assuntos
Apoptose , Cistationina/química , Retículo Endoplasmático/metabolismo , Homocistinúria/metabolismo , Lipídeos/química , Animais , Antibacterianos/farmacologia , Cistationina beta-Sintase/metabolismo , Células HEK293 , Células Hep G2 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Mutação , Necrose/patologia , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Desnaturação Proteica , Tunicamicina/farmacologia
8.
Am J Physiol Lung Cell Mol Physiol ; 305(1): L73-81, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23666751

RESUMO

Late-outgrowth endothelial colony-forming cells (ECFCs), a type of circulating endothelial progenitor cell (EPC), may contribute to pulmonary angiogenesis during development. Cord blood ECFCs from preterm newborns proliferate more rapidly than term ECFCs but are more susceptible to the adverse effects of hyperoxia. Recent studies suggest that bone marrow-derived EPCs protect against experimental lung injury via paracrine mechanisms independent of vascular engraftment. To determine whether human umbilical cord blood ECFCs from preterm and term newborns have therapeutic benefit in experimental neonatal lung injury, we isolated cord blood ECFCs from full-term and preterm newborns and prepared ECFC-conditioned medium (CM) to test its therapeutic benefit on fetal pulmonary artery endothelial cell (PAEC) proliferation and function as well as alveolar type 2 (AT2) cell growth. PAECs and AT2 cells were isolated from late-gestation fetal sheep. Additionally, we administered both ECFCs and ECFC-CM to bleomycin-exposed newborn rats, an experimental model of bronchopulmonary dysplasia (BPD). Both term ECFC-CM and preterm ECFC-CM promoted cell growth and angiogenesis in vitro. However, when ECFC-CM was collected during exposure to mild hyperoxia, the benefit of preterm ECFC-CM was no longer observed. In the bleomycin model of BPD, treatment with ECFC-CM (or CM from mature EC) effectively decreased right ventricular hypertrophy but had no effect on alveolar septation. We conclude that term ECFC-CM is beneficial both in vitro and in experimental BPD. During oxidative stress, preterm ECFC-CM, but not term ECFC-CM, loses its benefit. The inability of term ECFC-CM to promote alveolarization may limit its therapeutic potential.


Assuntos
Bleomicina/toxicidade , Displasia Broncopulmonar/complicações , Meios de Cultivo Condicionados/farmacologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Hipertensão Pulmonar/prevenção & controle , Neovascularização Fisiológica , Animais , Animais Recém-Nascidos , Antibióticos Antineoplásicos/toxicidade , Western Blotting , Displasia Broncopulmonar/induzido quimicamente , Proliferação de Células , Células Cultivadas , Feminino , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Hiperóxia , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/patologia , Técnicas Imunoenzimáticas , Recém-Nascido , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/metabolismo , Artéria Pulmonar/citologia , Artéria Pulmonar/metabolismo , Ratos , Ratos Sprague-Dawley
9.
Am J Physiol Heart Circ Physiol ; 304(12): H1624-33, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23585138

RESUMO

Cardiovascular disease risk factors, such as diabetes, hypertension, dyslipidemia, obesity, and physical inactivity, are all correlated with impaired endothelial nitric oxide synthase (eNOS) function and decreased nitric oxide (NO) production. NO-mediated regulation of mitochondrial biogenesis has been established in many tissues, yet the role of eNOS in vascular mitochondrial biogenesis and dynamics is unclear. We hypothesized that genetic eNOS deletion and 3-day nitric oxide synthase (NOS) inhibition in rodents would result in impaired mitochondrial biogenesis and defunct fission/fusion and autophagy profiles within the aorta. We observed a significant, eNOS expression-dependent decrease in mitochondrial electron transport chain (ETC) protein subunits from complexes I, II, III, and V in eNOS heterozygotes and eNOS null mice compared with age-matched controls. In response to NOS inhibition with NG-nitro-L-arginine methyl ester (L-NAME) treatment in Sprague Dawley rats, significant decreases were observed in ETC protein subunits from complexes I, III, and IV as well as voltage-dependent anion channel 1. Decreased protein content of upstream regulators of mitochondrial biogenesis, cAMP response element-binding protein and peroxisome proliferator-activated receptor-γ coactivator-1α, were observed in response to 3-day L-NAME treatment. Both genetic eNOS deletion and NOS inhibition resulted in decreased manganese superoxide dismutase protein. L-NAME treatment resulted in significant changes to mitochondrial dynamic protein profiles with decreased fusion, increased fission, and minimally perturbed autophagy. In addition, L-NAME treatment blocked mitochondrial adaptation to an exercise intervention in the aorta. These results suggest that eNOS/NO play a role in basal and adaptive mitochondrial biogenesis in the vasculature and regulation of mitochondrial turnover.


Assuntos
Adaptação Fisiológica , Endotélio Vascular/metabolismo , Mitocôndrias/metabolismo , Dinâmica Mitocondrial , Óxido Nítrico/metabolismo , Animais , Aorta/citologia , Aorta/metabolismo , Autofagia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Complexo de Proteínas da Cadeia de Transporte de Elétrons/genética , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Endotélio Vascular/citologia , Deleção de Genes , Expressão Gênica , Heterozigoto , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Ratos , Ratos Sprague-Dawley , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Canal de Ânion 1 Dependente de Voltagem/genética , Canal de Ânion 1 Dependente de Voltagem/metabolismo
10.
J Pediatr ; 163(3): 905-7, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23684109

RESUMO

Endothelial colony-forming cells (ECFCs) are decreased in the cord blood of preterm infants with moderate-to-severe bronchopulmonary dysplasia. We quantified ECFCs from infants with congenital diaphragmatic hernia, a neonatal disorder with severe lung hypoplasia. Unlike newborns who develop bronchopulmonary dysplasia, those with congenital diaphragmatic hernia had increased and highly-proliferative cord blood ECFCs.


Assuntos
Células Endoteliais , Sangue Fetal/citologia , Hérnias Diafragmáticas Congênitas , Células-Tronco , Proliferação de Células , Células Cultivadas , Células Endoteliais/fisiologia , Feminino , Hérnia Diafragmática/sangue , Humanos , Recém-Nascido , Contagem de Leucócitos , Leucócitos Mononucleares/fisiologia , Masculino , Células-Tronco/fisiologia
13.
Ann Am Thorac Soc ; 20(10): 1373-1388, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37772940

RESUMO

Despite growing recognition of the need for increased diversity among students, trainees, and faculty in health care, the medical workforce still lacks adequate representation from groups historically underrepresented in medicine (URiM). The subspecialty field of pediatric pulmonology is no exception. Although there have been efforts to address issues of diversity, equity, and inclusion (DEI) in our own field, gaps persist. To address these gaps, the members of the Diversity, Equity, and Inclusion Advisory Group (DEI-AG) of the American Thoracic Society Pediatrics Assembly created and distributed a Needs Assessment Survey in the United States and Canada to better understand the racial and ethnic demographics of the pediatric pulmonary workforce and to learn more about successes, gaps, and opportunities to enhance how we recruit, train, and retain a diverse workforce. The DEI-AG leadership cochairs convened a workshop to review the findings of the DEI Needs Assessment Survey and to develop strategies to improve the recruitment and retention of URiM fellows and faculty. This Official ATS Workshop Report aims to identify barriers and opportunities for recruitment, training, and career development within the field of pediatric pulmonology. Additionally, we offer useful strategies and resources to improve the recruitment of URiM residents, the mentorship of trainees and junior faculty, and the career development of URiM faculty in academic centers. This Workshop Report is an important first deliverable by the DEI-AG. We hope that this work, originating from within the Pediatrics Assembly, will serve as a model for other Assemblies, disciplines across the ATS, and other fields in Pediatrics.

14.
Am J Physiol Lung Cell Mol Physiol ; 302(9): L829-37, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22328358

RESUMO

Bronchopulmonary dysplasia (BPD) remains a major complication of prematurity resulting in significant morbidity and mortality. The pathology of BPD is multifactorial and leads to alveolar simplification and distal lung injury. Previous studies have shown a beneficial effect of systemic treatment with bone marrow-derived mesenchymal stromal cells (MSCs) and MSC-conditioned media (MSC-CM) leading to amelioration of the lung parenchymal and vascular injury in vivo in the hyperoxia murine model of BPD. It is possible that the beneficial response from the MSCs is at least in part due to activation of endogenous lung epithelial stem cells. Bronchioalveolar stem cells (BASCs) are an adult lung stem cell population capable of self-renewal and differentiation in culture, and BASCs proliferate in response to bronchiolar and alveolar lung injury in vivo. Systemic treatment of neonatal hyperoxia-exposed mice with MSCs or MSC-CM led to a significant increase in BASCs compared with untreated controls. Treatment of BASCs with MSC-CM in culture showed an increase in growth efficiency, indicating a direct effect of MSCs on BASCs. Lineage tracing data in bleomycin-treated adult mice showed that Clara cell secretory protein-expressing cells including BASCs are capable of contributing to alveolar repair after lung injury. MSCs and MSC-derived factors may stimulate BASCs to play a role in the repair of alveolar lung injury found in BPD and in the restoration of distal lung cell epithelia. This work highlights the potential important role of endogenous lung stem cells in the repair of chronic lung diseases.


Assuntos
Células-Tronco Adultas/patologia , Displasia Broncopulmonar/terapia , Hiperóxia/terapia , Transplante de Células-Tronco Mesenquimais , Animais , Animais Recém-Nascidos , Bleomicina , Bronquíolos/patologia , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patologia , Contagem de Células , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Modelos Animais de Doenças , Humanos , Hiperóxia/metabolismo , Hiperóxia/patologia , Recém-Nascido , Peptídeos e Proteínas de Sinalização Intercelular , Pulmão/metabolismo , Pulmão/patologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/metabolismo , Lesão Pulmonar/patologia , Camundongos , Peptídeos/metabolismo , Proteína C Associada a Surfactante Pulmonar , Mucosa Respiratória/patologia , Uteroglobina/metabolismo
15.
Eur Respir J ; 40(6): 1516-22, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22496315

RESUMO

Bronchopulmonary dysplasia (BPD), the chronic lung disease of prematurity, is associated with impaired vascular and alveolar growth. Antenatal factors contribute to the risk for developing BPD by unclear mechanisms. Endothelial progenitor cells, such as angiogenic circulating progenitor cells (CPCs) and late-outgrowth endothelial colony-forming cells (ECFCs), may contribute to angiogenesis in the developing lung. We hypothesise that cord blood angiogenic CPCs and ECFCs are decreased in preterm infants with moderate and severe BPD. We quantified ECFCs and the CPC/nonangiogenic-CPC ratio (CPC/non-CPC) in cord blood samples from 62 preterm infants and assessed their relationships to maternal and perinatal risk factors as well as BPD severity. The CPC/non-CPC ratio and ECFC number were compared between preterm infants with mild or no BPD and those with moderate or severe BPD. ECFC number (p<0.001) and CPC/non-CPC ratio (p<0.05) were significantly decreased in cord blood samples of preterm infants who subsequently developed moderate or severe BPD. Gestational age and birth weight were not associated with either angiogenic marker. Circulating vascular progenitor cells are decreased in the cord blood of preterm infants who develop moderate and severe BPD. These findings suggest that prenatal factors contribute to late respiratory outcomes in preterm infants.


Assuntos
Displasia Broncopulmonar/sangue , Displasia Broncopulmonar/patologia , Sangue Fetal/citologia , Células-Tronco/citologia , Células Endoteliais/citologia , Feminino , Citometria de Fluxo/métodos , Humanos , Recém-Nascido , Leucócitos Mononucleares/citologia , Masculino , Exposição Materna , Neovascularização Fisiológica , Gravidez , Risco , Fatores de Risco , Resultado do Tratamento
17.
Am J Physiol Lung Cell Mol Physiol ; 299(6): L735-48, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20709730

RESUMO

To determine the separate and interactive effects of fetal inflammation and neonatal hyperoxia on the developing lung, we hypothesized that: 1) antenatal endotoxin (ETX) causes sustained abnormalities of infant lung structure; and 2) postnatal hyperoxia augments the adverse effects of antenatal ETX on infant lung growth. Escherichia coli ETX or saline (SA) was injected into amniotic sacs in pregnant Sprague-Dawley rats at 20 days of gestation. Pups were delivered 2 days later and raised in room air (RA) or moderate hyperoxia (O2, 80% O2 at Denver's altitude, ∼65% O2 at sea level) from birth through 14 days of age. Heart and lung tissues were harvested for measurements. Intra-amniotic ETX caused right ventricular hypertrophy (RVH) and decreased lung vascular endothelial growth factor (VEGF) and VEGF receptor-2 (VEGFR-2) protein contents at birth. In ETX-exposed rats (ETX-RA), alveolarization and vessel density were decreased, pulmonary vascular wall thickness percentage was increased, and RVH was persistent throughout the study period compared with controls (SA-RA). After antenatal ETX, moderate hyperoxia increased lung VEGF and VEGFR-2 protein contents in ETX-O2 rats and improved their alveolar and vascular structure and RVH compared with ETX-RA rats. In contrast, severe hyperoxia (≥95% O2 at Denver's altitude) further reduced lung vessel density after intra-amniotic ETX exposure. We conclude that intra-amniotic ETX induces fetal pulmonary hypertension and causes persistent abnormalities of lung structure with sustained pulmonary hypertension in infant rats. Moreover, moderate postnatal hyperoxia after antenatal ETX restores lung growth and prevents pulmonary hypertension during infancy.


Assuntos
Animais Recém-Nascidos , Endotoxinas/farmacologia , Feto/efeitos dos fármacos , Hiperóxia , Hipertensão Pulmonar/induzido quimicamente , Pulmão/efeitos dos fármacos , Pulmão/crescimento & desenvolvimento , Animais , Feminino , Feto/anatomia & histologia , Idade Gestacional , Humanos , Hipertensão Pulmonar/fisiopatologia , Hipertrofia Ventricular Direita/induzido quimicamente , Lactente , Pulmão/anatomia & histologia , Pulmão/fisiopatologia , Oxigênio/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos , Ratos Sprague-Dawley
18.
Am J Physiol Lung Cell Mol Physiol ; 298(3): L315-23, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20008116

RESUMO

Neonatal hyperoxia impairs vascular and alveolar growth in mice and decreases endothelial progenitor cells. To determine the role of bone marrow-derived cells in restoration of neonatal lung structure after injury, we studied a novel bone marrow myeloid progenitor cell population from Tie2-green fluorescent protein (GFP) transgenic mice (bone marrow-derived angiogenic cells; BMDAC). We hypothesized that treatment with BMDAC would restore normal lung structure in infant mice during recovery from neonatal hyperoxia. Neonatal mice (1-day-old) were exposed to 80% oxygen for 10 days. BMDACs (1 x 10(5)), embryonic endothelial progenitor cells, mouse embryonic fibroblasts (control), or saline were then injected into the pulmonary circulation. At 21 days of age, saline-treated mice had enlarged alveoli, reduced septation, and a reduction in vascular density. In contrast, mice treated with BMDAC had complete restoration of lung structure that was indistinguishable from room air controls. BMDAC comprised 12% of distal lung cells localized to pulmonary vessels or alveolar type II (AT2) cells and persist (8.8%) for 8 wk postinjection. Coculture of AT2 cells or lung endothelial cells (luEC) with BMDAC augmented AT2 and luEC cell growth in vitro. We conclude that treatment with BMDAC after neonatal hyperoxia restores lung structure in this model of bronchopulmonary dysplasia.


Assuntos
Células da Medula Óssea/citologia , Células Endoteliais/citologia , Hiperóxia/patologia , Neovascularização Fisiológica , Alvéolos Pulmonares/irrigação sanguínea , Alvéolos Pulmonares/patologia , Animais , Animais Recém-Nascidos , Proliferação de Células , Ensaio de Unidades Formadoras de Colônias , Células Endoteliais/transplante , Citometria de Fluxo , Imunofluorescência , Camundongos , Fenótipo , Fatores de Tempo
19.
Am J Respir Crit Care Med ; 180(5): 454-61, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-19483112

RESUMO

RATIONALE: Preterm birth and hyperoxic exposure increase the risk for bronchopulmonary dysplasia (BPD), a chronic lung disease characterized by impaired vascular and alveolar growth. Endothelial progenitor cells, such as self-renewing highly proliferative endothelial colony-forming cells (ECFCs), may participate in vascular repair. The effect of hyperoxia on ECFC growth is unknown. OBJECTIVES: We hypothesize that umbilical cord blood (CB) from premature infants contains more ECFCs with greater growth potential than term CB. However, preterm ECFCs may be more susceptible to hyperoxia. METHODS: ECFC colonies were quantified by established methods and characterized by immunohistochemistry and flow cytometry. Growth kinetics were assessed in room air and hyperoxia (FI(O(2)) = 0.4). MEASUREMENTS AND MAIN RESULTS: Preterm CB (28-35 wk gestation) yielded significantly more ECFC colonies than term CB. Importantly, we found that CD45(-)/CD34(+)/CD133(+)/VEGFR-2(+) cell number did not correlate with ECFC colony count. Preterm ECFCs demonstrated increased growth compared with term ECFCs. Hyperoxia impaired growth of preterm but not term ECFCs. Treatment with superoxide dismutase and catalase enhanced preterm ECFC growth during hyperoxia. CONCLUSIONS: Preterm ECFCs appear in increased numbers and proliferate more rapidly but have an increased susceptibility to hyperoxia compared with term ECFCs. Antioxidants protect preterm ECFCs from hyperoxia.


Assuntos
Células Endoteliais/metabolismo , Hiperóxia/sangue , Recém-Nascido Prematuro/sangue , Células-Tronco/metabolismo , Antioxidantes/uso terapêutico , Células Endoteliais/efeitos dos fármacos , Sangue Fetal/metabolismo , Humanos , Recém-Nascido , Recém-Nascido Prematuro/crescimento & desenvolvimento , Células-Tronco/efeitos dos fármacos
20.
Int J Pediatr Otorhinolaryngol ; 138: 110355, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33152958

RESUMO

Foreign body (FB) aspiration is potentially life-threatening in children. A variety of sources and objects have been noted in aspiration events with possible complications ranging from mild to life-threatening. While rare, barium aspiration can cause severe complications, and removal is particularly challenging. Complications of retained barium include acute respiratory distress syndrome, pneumonitis, sepsis, even death. Regardless of the foreign body's identity, substance removal is critical in management. Resourcefulness of removal techniques and an interdisciplinary approach may allow for maximally effective management. We present a case of a pediatric barium aspiration from an unknown source and review evaluation and management strategies.


Assuntos
Bário/efeitos adversos , Corpos Estranhos , Aspiração Respiratória , Brônquios , Broncoscopia , Pré-Escolar , Corpos Estranhos/diagnóstico por imagem , Humanos , Masculino , Aspiração Respiratória/diagnóstico por imagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA