Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Respir Physiol Neurobiol ; 313: 104060, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37031925

RESUMO

DNA methylation is necessary for developmental gene regulation, but adverse environments result in aberrant methylation and gene silencing. The current pilot study tested the hypothesis that treatment with DNA methylation inhibitors (decitabine; RG108) would improve alveolarization in a newborn murine model of severe bronchopulmonary dysplasia. Newborn mice exposed to maternal inflammation (LPS) and neonatal hyperoxia (85% O2) were treated with decitabine (p3, 0.1 mg/kg; p2, 4, 6, 0.1 mg/kg; or p2, 4, 6, 0.15 mg/kg) or RG108 (p3, 0.0013 mg/kg) delivered intranasally. Modest improvements in alveolarization were observed with decitabine, but no differences were observed with RG108. Attenuated phospho-SMAD2/3 levels and greater surfactant protein C protein levels compared to vehicle were observed with some tested doses. No detrimental side effects were observed with the doses used in this study. In summary, our pilot investigations identified a safe dose for intranasal administration of both methylation inhibitors and provides a foundation for further studies into methylation inhibitors in the context of neonatal lung injury.


Assuntos
Displasia Broncopulmonar , Hiperóxia , Animais , Camundongos , Animais Recém-Nascidos , Decitabina/farmacologia , Decitabina/uso terapêutico , Decitabina/metabolismo , Modelos Animais de Doenças , DNA/metabolismo , DNA/farmacologia , DNA/uso terapêutico , Hiperóxia/metabolismo , Pulmão/metabolismo , Projetos Piloto
2.
Redox Biol ; 38: 101783, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33202301

RESUMO

Preterm birth is still a major health problem and maternal inflammation has been shown to play a role. The combination of maternal inflammation and neonatal hyperoxia contributes to epigenetic changes that influence gene expression and the development of bronchopulmonary dysplasia (BPD). We have previously demonstrated suppression of miR-29b and increases in DNA methylation in infants with severe BPD and in our mouse model of maternal inflammation and neonatal hyperoxia exposure. The present studies further explored epigenetic changes in the murine model to include histone methylation. We identified a global suppression of histone methylation in exposed mice and validated decreases in expression in well-defined histone modifications, specifically H3K4me3, H3K27me3, H3K36me2, H3K79me2, and H4K20me3. We further tested the hypothesis that restoration of miR-29b expression would restore the histone methylation marks. Using lipid nanoparticle delivery of miR-29b, partial to full methylation was reestablished for H3K4me3, H3K27me3, and H4K20me3; all tri-methylation marks. To identify the causes of decreased methylation in exposed mice, we measured commonly identified methylases and demethylases. We found a decreased expression of SUV40H2, a methylase primarily associated with H4K20me3. Further studies are needed to identify the causes for the decreased global histone methylation and potential therapeutic opportunities.


Assuntos
MicroRNAs , Nascimento Prematuro , Animais , Metilação de DNA , Suplementos Nutricionais , Feminino , Histonas/metabolismo , Inflamação/genética , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Gravidez
3.
Am J Physiol Lung Cell Mol Physiol ; 317(2): L167-L174, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31042079

RESUMO

The effects of maternal obesity on lung development have been recognized, and speculation is that these diseases are not simply because of accelerated pulmonary decline with aging but with a failure to achieve optimal lung development during early life. These studies tested the hypothesis that maternal obesity alters signaling pathways during the course of lung development that may affect life-long pulmonary health. Adult female mice were fed 60% fat [high-fat diet (HFD)] or 10% fat [control diet (CD)] for 8 wk before mating and through weaning. Pup lung tissues were collected at postnatal days (PN) 7, 21, and 90 (after receiving HFD or CD as adults). At PN7, body weights from HFD were greater than CD but lung weight-to-body weight ratios were lower. In lung tissues, NFκB-mediated inflammation was greater in HFD pups at PN21 and phospho-/total STAT3, phospho-/total VEGF receptor 2, and total AKT protein levels were lower with maternal HFD and protein tyrosine phosphatase B1 levels were increased. Decreased platelet endothelial cell adhesion molecule levels were observed at PN21 and at PN90 in the pups exposed to maternal HFD. Morphometry indicated that the pups exposed to maternal or adult HFD had fewer alveoli, and the effect was additive. Decreases in pulmonary resistance, elastance, and compliance were observed because of adult HFD diet and decreases in airway resistance and increases in inspiratory capacity because of maternal HFD. In conclusion, maternal HFD disrupts signaling pathways in the early developing lung and may contribute to deficiencies in lung function and increased susceptibility in adults.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Pulmão/crescimento & desenvolvimento , Obesidade/etiologia , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Animais , Animais Recém-Nascidos , Feminino , Inflamação/complicações , Pulmão/efeitos dos fármacos , Masculino , Camundongos , Gravidez , Desmame
4.
Reprod Sci ; 26(6): 757-765, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30092744

RESUMO

Accreta and gestational trophoblastic disease (ie, choriocarcinoma) are placental pathologies characterized by hyperproliferative and invasive trophoblasts. Cellular proliferation, migration, and invasion are heavily controlled by actin-binding protein (ABP)-mediated actin dynamics. The ABP vasodilator-stimulated phosphoprotein (VASP) carries key regulatory role. Profilin-1, cofilin-1, and VASP phosphorylated at Ser157 (pVASP-S157) and Ser239 (pVASP-S239) are ABPs that regulate actin polymerization and stabilization and facilitate cell metastases. Docosahexaenoic acid (DHA) inhibits cancer cell migration and proliferation. We hypothesized that analogous to malignant cells, ABPs regulate these processes in extravillous trophoblasts (EVTs), which exhibit aberrant expression in placenta accreta. Placental-myometrial junction biopsies of histologically confirmed placenta accreta had significantly increased immunostaining levels of cofilin-1, VASP, pVASP-S239, and F-actin. Treatment of choriocarcinoma-derived trophoblast (BeWo) cells with DHA (30 µM) for 24 hours significantly suppressed proliferation, migration, and pVASP-S239 levels and altered protein profiles consistent with increased apoptosis. We concluded that in accreta changes in the ABP expression profile were a response to restore homeostasis by counteracting the hyperproliferative and invasive phenotype of the EVT. The observed association between VASP phosphorylation, apoptosis, and trophoblast proliferation and migration suggest that DHA may offer a therapeutic solution for conditions where EVT is hyperinvasive.


Assuntos
Moléculas de Adesão Celular/metabolismo , Cofilina 1/análise , Ácidos Docosa-Hexaenoicos/farmacologia , Proteínas dos Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Placenta Acreta/metabolismo , Profilinas/análise , Trofoblastos/fisiologia , Apoptose/efeitos dos fármacos , Moléculas de Adesão Celular/análise , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Coriocarcinoma , Feminino , Humanos , Proteínas dos Microfilamentos/análise , Fosfoproteínas/análise , Fosforilação/efeitos dos fármacos , Placenta/química , Gravidez , Trofoblastos/efeitos dos fármacos
5.
Respir Res ; 19(1): 229, 2018 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-30463566

RESUMO

BACKGROUND: Hyperoxia is a frequently employed therapy for prematurely born infants, induces lung injury and contributes to development of bronchopulmonary dysplasia (BPD). BPD is characterized by decreased cellular proliferation, cellular migration, and failure of injury repair systems. Actin binding proteins (ABPs) such as VASP, cofilin1, and profilin1 regulate cell proliferation and migration via modulation of actin dynamics. Lung mesenchymal stem cells (L-MSCs) initiate repair processes by proliferating, migrating, and localizing to sites of injury. These processes have not been extensively explored in hyperoxia induced lung injury and repair. METHODS: ABPs and CD146+ L-MSCs were analyzed by immunofluorescence in human lung autopsy tissues from infants with and without BPD and by western blot in lung tissue homogenates obtained from our murine model of newborn hyperoxic lung injury. RESULTS: Decreased F-actin content, ratio of VASPpS157/VASPpS239, and profilin 1 expression were observed in human lung tissues but this same pattern was not observed in lungs from hyperoxia-exposed newborn mice. Increases in cofilin1 expression were observed in both human and mouse tissues at 7d indicating a dysregulation in actin dynamics which may be related to altered growth. CD146 levels were elevated in human and newborn mice tissues (7d). CONCLUSION: Altered phosphorylation of VASP and expression of profilin 1 and cofilin 1 in human tissues indicate that the pathophysiology of BPD involves dysregulation of actin binding proteins. Lack of similar changes in a mouse model of hyperoxia exposure imply that disruption in actin binding protein expression may be linked to interventions or morbidities other than hyperoxia alone.


Assuntos
Displasia Broncopulmonar/metabolismo , Moléculas de Adesão Celular/metabolismo , Cofilina 1/biossíntese , Hiperóxia/metabolismo , Lesão Pulmonar/metabolismo , Proteínas dos Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Profilinas/biossíntese , Animais , Animais Recém-Nascidos , Displasia Broncopulmonar/patologia , Cofilina 1/genética , Feminino , Expressão Gênica , Humanos , Hiperóxia/patologia , Recém-Nascido , Lesão Pulmonar/patologia , Camundongos , Camundongos Endogâmicos C3H , Fosforilação/fisiologia , Gravidez , Profilinas/genética , Distribuição Aleatória
6.
Clin Exp Pharmacol Physiol ; 45(6): 556-562, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29266319

RESUMO

The L-arginine/NO pathway is an important regulator of pulmonary hypertension, the leading cause of mortality in patients with the chronic lung disease of prematurity, bronchopulmonary dysplasia. L-arginine can be metabolized by NO synthase (NOS) to form L-citrulline and NO, a potent vasodilator. Alternatively, L-arginine can be metabolized by arginase to form urea and L-ornithine, a precursor to collagen and proline formation important in vascular remodelling. In the current study, we hypothesized that C3H/HeN mice exposed to prolonged hyperoxia would have increased arginase expression and pulmonary vascular wall cell proliferation. C3H/HeN mice were exposed to 14 days of 85% O2 or room air and lung homogenates analyzed by western blot for protein levels of arginase I, arginase II, endothelial NOS (eNOS), ornithine decarboxylase (ODC), ornithine aminotransferase (OAT), and α-smooth muscle actin (α-SMA). Hyperoxia did not change arginase I or eNOS protein levels. However, arginase II protein levels were 15-fold greater after hyperoxia exposure than in lungs exposed to room air. Greater protein levels of ODC and OAT were found in lungs following hyperoxic exposure than in room air animals. α-SMA protein levels were found to be 7-fold greater in the hyperoxia exposed lungs than in room air lungs. In the hyperoxia exposed lungs there was evidence of greater pulmonary vascular wall cell proliferation by α-SMA immunohistochemistry than in room air lungs. Taken together, these data are consistent with a more proliferative vascular phenotype, and may explain the propensity of patients with bronchopulmonary dysplasia to develop pulmonary hypertension.


Assuntos
Actinas/metabolismo , Arginase/biossíntese , Displasia Broncopulmonar/complicações , Displasia Broncopulmonar/metabolismo , Hiperóxia/complicações , Animais , Displasia Broncopulmonar/patologia , Proliferação de Células , Modelos Animais de Doenças , Indução Enzimática , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Ornitina Descarboxilase/metabolismo , Ornitina-Oxo-Ácido Transaminase/metabolismo
7.
Am J Physiol Heart Circ Physiol ; 314(3): H573-H579, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29212791

RESUMO

Sex-related differences in cardiovascular health and disease have been identified, with males having a higher incidence of cardiovascular events but females more likely to develop arrhythmias. Adverse fetal environments are now accepted as a cause for the development of cardiovascular diseases in adulthood, but sex-related differences in response to adverse fetal environments have not been extensively explored. The combination of both in utero and postnatal exposure to inflammation is highly relevant for the infant that is born preterm or has clinical complications at birth or in early postnatal life. We have previously observed cardiac contractile deficiencies and dysregulation of Ca2+-handling proteins in our model of maternal lipopolysaccharide (LPS) and neonatal hyperoxia exposures (LPS/O2). This investigation tested the hypothesis that there are sex-related differences in the adult pathologies after exposure to perinatal inflammation. Using pressure-volume assessments, males exposed to LPS/O2 had more pronounced contractile deficiencies than similarly exposed females, but females tended to have long PR intervals. While both sexes demonstrated decreases in α-myosin heavy chain and connexin 43 after LPS/O2 exposure compared with saline/room air controls, females indicated aberrant increases in microRNA 208a, microRNA 208b, and desmin expression. Our study supports our hypothesis that early life exposure to inflammation results in sex-dependent deficits in cardiovascular function. NEW & NOTEWORTHY Sex-specific differences in cardiovascular disease are recognized, but the mechanisms and origins are not well understood. Adverse maternal environments can influence cardiac development and later cardiovascular disease. This study identifies sex-dependent differences in cardiac disease associated with perinatal inflammation.


Assuntos
Cardiomiopatias/etiologia , Inflamação/complicações , Complicações na Gravidez , Efeitos Tardios da Exposição Pré-Natal , Animais , Animais Recém-Nascidos , Cardiomiopatias/metabolismo , Cardiomiopatias/fisiopatologia , Conexina 43/metabolismo , Desmina/metabolismo , Modelos Animais de Doenças , Feminino , Frequência Cardíaca , Hiperóxia/complicações , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/fisiopatologia , Lipopolissacarídeos , Masculino , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Contração Miocárdica , Miocárdio/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Gravidez , Complicações na Gravidez/induzido quimicamente , Complicações na Gravidez/metabolismo , Complicações na Gravidez/fisiopatologia , Fatores de Risco , Fatores Sexuais , Função Ventricular Esquerda , Pressão Ventricular
8.
Am J Physiol Lung Cell Mol Physiol ; 313(2): L339-L349, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28473324

RESUMO

Even with advances in the care of preterm infants, chronic lung disease or bronchopulmonary dysplasia (BPD) continues to be a significant pulmonary complication. Among those diagnosed with BPD, a subset of infants develop severe BPD with disproportionate pulmonary morbidities. In addition to decreased alveolarization, these infants develop obstructive and/or restrictive lung function due to increases in or dysregulation of extracellular matrix proteins. Analyses of plasma obtained from preterm infants during the first week of life indicate that circulating miR-29b is suppressed in infants that subsequently develop BPD and that decreased circulating miR-29b is inversely correlated with BPD severity. Our mouse model mimics the pathophysiology observed in infants with severe BPD, and we have previously reported decreased pulmonary miR-29b expression in this model. The current studies tested the hypothesis that adeno-associated 9 (AAV9)-mediated restoration of miR-29b in the developing lung will improve lung alveolarization and minimize the deleterious changes in matrix deposition. Pregnant C3H/HeN mice received an intraperitoneal LPS injection on embryonic day 16 and newborn pups were exposed to 85% oxygen from birth to 14 days of life. On postnatal day 3, AAV9-miR-29b or AAV9-control was administered intranasally. Mouse lung tissues were then analyzed for changes in miR-29 expression, alveolarization, and matrix protein levels and localization. Although only modest improvements in alveolarization were detected in the AAV9-miR29b-treated mice at postnatal day 28, treatment completely attenuated defects in matrix protein expression and localization. Our data suggest that miR-29b restoration may be one component of a novel therapeutic strategy to treat or prevent severe BPD in prematurely born infants.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Hiperóxia/metabolismo , Inflamação/metabolismo , MicroRNAs/metabolismo , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Humanos , Recém-Nascido , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Oxigênio/administração & dosagem
9.
Mol Cancer Ther ; 15(9): 2220-31, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27496138

RESUMO

Profilin 1, cofilin 1, and vasodialator-stimulated phosphoprotein (VASP) are actin-binding proteins (ABP) that regulate actin remodeling and facilitate cancer cell metastases. miR-17-92 is highly expressed in metastatic tumors and profilin1 and cofilin1 are predicted targets. Docosahexaenoic acid (DHA) inhibits cancer cell proliferation and adhesion. These studies tested the hypothesis that the metastatic phenotype is driven by changes in ABPs including alternative phosphorylation and/or changes in subcellular localization. In addition, we tested the efficacy of DHA supplementation to attenuate or inhibit these changes. Human lung cancer tissue sections were analyzed for F-actin content and expression and cellular localization of profilin1, cofilin1, and VASP (S157 or S239 phosphorylation). The metastatic phenotype was investigated in A549 and MLE12 cells lines using 8 Br-cAMP as a metastasis inducer and DHA as a therapeutic agent. Migration was assessed by wound assay and expression measured by Western blot and confocal analysis. miR-17-92 expression was measured by qRT-PCR. Results indicated increased expression and altered cellular distribution of profilin1/VASP(pS157), but no changes in cofilin1/VASP(pS239) in the human malignant tissues compared with normal tissues. In A549 and MLE12 cells, the expression patterns of profilin1/VASP(pS157) or cofilin1/VASP(pS239) suggested an interaction in regulation of actin dynamics. Furthermore, DHA inhibited cancer cell migration and viability, ABP expression and cellular localization, and modulated expression of miR-17-92 in A549 cells with minimal effects in MLE12 cells. Further investigations are warranted to understand ABP interactions, changes in cellular localization, regulation by miR-17-92, and DHA as a novel therapeutic. Mol Cancer Ther; 15(9); 2220-31. ©2016 AACR.


Assuntos
Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Cofilina 1/genética , Cofilina 1/metabolismo , Expressão Gênica , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Profilinas/genética , Profilinas/metabolismo , Apoptose/genética , Biomarcadores , Linhagem Celular Tumoral , Movimento Celular/genética , Núcleo Celular/metabolismo , AMP Cíclico/metabolismo , Desidroepiandrosterona/administração & dosagem , Suplementos Nutricionais , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , MicroRNAs/genética , Metástase Neoplásica , Fosforilação/efeitos dos fármacos , Ligação Proteica , Transporte Proteico/efeitos dos fármacos
10.
Life Sci ; 155: 1-9, 2016 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-27157519

RESUMO

AIMS: Deaths associated with cancer metastasis have steadily increased making the need for newer, anti-metastatic therapeutics imparative. Gelsolin and vimentin, actin binding proteins expressed in metastatic tumors, participate in actin remodelling and regulate cell migration. Docosahexaenoic acid (DHA) limits cancer cell proliferation and adhesion but the mechanisms involved in reducing metastatic phenotypes are unknown. We aimed to investigate the effects of DHA on gelsolin and vimentin expression, and ultimately cell migration and proliferation, in this context. MAIN METHODS: Non-invasive lung epithelial cells (MLE12) and invasive lung cancer cells (A549) were treated with DHA (30µmol/ml) or/and 8 bromo-cyclic adenosine monophosphate (8 Br-cAMP) (300µmol/ml) for 6 or 24h either before (pre-treatment) or after (post-treatment) plating in transwells. Migration was assessed by the number of cells that progressed through the transwell. Gelsolin and vimentin expression were measured by Western blot and confocal microscopy in cells, and by immunohistochemistry in human lung cancer biopsy samples. KEY FINDINGS: A significant decrease in cell migration was detected for A549 cells treated with DHA verses control but this same decrease was not seen in MLE12 cells. DHA and 8 Br-cAMP altered gelsolin and vimentin expression but no clear pattern of change was observed. Immunofluorescence staining indicated slightly higher vimentin expression in human lung tissue that was malignant compared to control. SIGNIFICANCE: Collectively, our data indicate that DHA inhibits cancer cell migration and further suggests that vimentin and gelsolin may play secondary roles in cancer cell migration and proliferation, but are not the primary regulators.


Assuntos
Movimento Celular/efeitos dos fármacos , Ácidos Docosa-Hexaenoicos/farmacologia , Gelsolina/farmacologia , Neoplasias Pulmonares/patologia , Vimentina/metabolismo , Células A549 , Humanos , Neoplasias Pulmonares/metabolismo
11.
Sci Rep ; 6: 22276, 2016 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-26940787

RESUMO

Persistent macrophages were observed in the lungs of murine offspring exposed to maternal LPS and neonatal hyperoxia. Maternal docosahexaenoic acid (DHA) supplementation prevented the accumulation of macrophages and improved lung development. We hypothesized that these macrophages are responsible for pathologies observed in this model and the effects of DHA supplementation. Primary macrophages were isolated from adult mice fed standard chow, control diets, or DHA supplemented diets. Macrophages were exposed to hyperoxia (O2) for 24 h and LPS for 6 h or 24 h. Our data demonstrate significant attenuation of Notch 1 and Jagged 1 protein levels in response to DHA supplementation in vivo but similar results were not evident in macrophages isolated from mice fed standard chow and supplemented with DHA in vitro. Co-culture of activated macrophages with MLE12 epithelial cells resulted in the release of high mobility group box 1 and leukotriene B4 from the epithelial cells and this release was attenuated by DHA supplementation. Collectively, our data indicate that long term supplementation with DHA as observed in vivo, resulted in deceased Notch 1/Jagged 1 protein expression however, DHA supplementation in vitro was sufficient to suppress release LTB4 and to protect epithelial cells in co-culture.


Assuntos
Ácidos Docosa-Hexaenoicos/administração & dosagem , Células Epiteliais/fisiologia , Hiperóxia/prevenção & controle , Imunossupressores/administração & dosagem , Proteína Jagged-1/metabolismo , Macrófagos/efeitos dos fármacos , Receptor Notch1/metabolismo , Animais , Células Cultivadas , Técnicas de Cocultura , Ácidos Docosa-Hexaenoicos/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Hiperóxia/imunologia , Imunossupressores/farmacologia , Mediadores da Inflamação/metabolismo , Proteína Jagged-1/genética , Pulmão/fisiologia , Macrófagos/imunologia , Masculino , Exposição Materna/efeitos adversos , Camundongos , Camundongos Endogâmicos C3H , Receptor Notch1/genética , Transdução de Sinais/efeitos dos fármacos
12.
Am J Physiol Lung Cell Mol Physiol ; 309(5): L441-8, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26138643

RESUMO

We have previously shown that an adverse perinatal environment significantly alters lung growth and development and results in persistently altered cardiopulmonary physiology in adulthood. Our model of maternal LPS treatment followed by 14 days of neonatal hyperoxia exposure causes severe pulmonary disease characterized by permanent decreases in alveolarization and diffuse interstitial fibrosis. The current investigations tested the hypothesis that dysregulation of Notch signaling pathways contributes to the permanently altered lung phenotype in our model and that the improvements we have observed previously with maternal docosahexaenoic acid (DHA) supplementation are mediated through normalization of Notch-related protein expression. Results indicated that inflammation (IL-6 levels) and oxidation (F2a-isoprostanes) persisted through 8 wk of life in mice exposed to LPS/O2 perinatally. These changes were attenuated by maternal DHA supplementation. Modest but inconsistent differences were observed in Notch-pathway proteins Jagged 1, DLL 1, PEN2, and presenilin-2. We detected substantial increases in markers of apoptosis including PARP-1, APAF-1, caspase-9, BCL2, and HMGB1, and these increases were attenuated in mice that were nursed by DHA-supplemented dams during the perinatal period. Although Notch signaling is not significantly altered at 8 wk of age in mice with perinatal exposure to LPS/O2, our findings indicate that persistent apoptosis continues to occur at 8 wk of age. We speculate that ongoing apoptosis may contribute to persistently altered lung development and may further enhance susceptibility to additional pulmonary disease. Finally, we found that maternal DHA supplementation prevented sustained inflammation, oxidation, and apoptosis in our model.


Assuntos
Apoptose/efeitos dos fármacos , Ácidos Docosa-Hexaenoicos/farmacologia , Inflamação/tratamento farmacológico , Pneumopatias/tratamento farmacológico , Pulmão/patologia , Receptores Notch/metabolismo , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Hipóxia Celular/fisiologia , Suplementos Nutricionais , Modelos Animais de Doenças , Ácidos Docosa-Hexaenoicos/uso terapêutico , Feminino , Proteína HMGB1/metabolismo , Hiperóxia/patologia , Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interleucina-6/metabolismo , Proteína Jagged-1 , Lipopolissacarídeos , Pneumopatias/patologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Estresse Oxidativo/efeitos dos fármacos , Presenilina-1/metabolismo , Presenilina-2/metabolismo , Proteínas Serrate-Jagged , Transdução de Sinais/efeitos dos fármacos
13.
J Nutr ; 144(3): 258-66, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24453131

RESUMO

The preterm infant is often exposed to maternal and neonatal inflammatory stimuli and is born with immature lungs, resulting in a need for oxygen therapy. Nutritional intervention with docosahexaenoic acid (DHA; 6.3 g/kg of diet) has been shown to attenuate inflammation in various human diseases. Previous studies demonstrated that maternal DHA supplementation during late gestation and lactation attenuated hyperoxic lung injury in newborn mouse pups. In the present studies, we tested the hypothesis that DHA supplementation to the dam would reduce hyperoxic lung injury and growth deficits in a more severe model of systemic maternal inflammation, including lipopolysaccharide (LPS) and neonatal hyperoxia exposure. On embryonic day 16, dams were placed on DHA (6.3 g DHA/kg diet) or control diets and injected with saline or LPS. Diets were maintained through weaning. At birth, pups were placed in room air or hyperoxia for 14 d. Improvements in birth weight (P < 0.01), alveolarization (P ≤ 0.01), and pulmonary function (P ≤ 0.03) at 2 and 8 wk of age were observed in pups exposed to perinatal inflammation and born to DHA-supplemented dams compared with control diet-exposed pups. These improvements were associated with decreases in tissue macrophage numbers (P < 0.01), monocyte chemoattractant protein-1 expression (P ≤ 0.05), and decreases in soluble receptor for advanced glycation end products concentrations (P < 0.01) at 2 and 8 wk. Furthermore, DHA supplementation attenuated pulmonary fibrosis, which was associated with the reduction of matrix metalloproteinases 2, 3, and 8 (P ≤ 0.03) and collagen mRNA (P ≤ 0.05), and decreased collagen (P < 0.01) and vimentin (P ≤ 0.03) protein concentrations. In a model of severe inflammation, maternal DHA supplementation lessened inflammation and improved lung growth in the offspring. Maternal supplementation with DHA may be a therapeutic strategy to reduce neonatal inflammation.


Assuntos
Suplementos Nutricionais , Ácidos Docosa-Hexaenoicos/administração & dosagem , Retardo do Crescimento Fetal/tratamento farmacológico , Inflamação/tratamento farmacológico , Pulmão/efeitos dos fármacos , Fenômenos Fisiológicos da Nutrição Materna , Animais , Animais Recém-Nascidos , Peso ao Nascer , Quimiocina CCL2/metabolismo , Dieta , Modelos Animais de Doenças , Feminino , Desenvolvimento Fetal/efeitos dos fármacos , Fibrose/tratamento farmacológico , Fibrose/metabolismo , Hiperóxia/tratamento farmacológico , Lipopolissacarídeos/efeitos adversos , Pulmão/metabolismo , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 3 da Matriz/metabolismo , Metaloproteinase 8 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Fosforilação , RNA Mensageiro/metabolismo , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo
14.
Life Sci ; 94(2): 164-71, 2014 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-24291255

RESUMO

AIMS: Maternal inflammation is a risk factor for preterm birth, and premature infants are often exposed to supplemental oxygen as a life-sustaining therapy. While more immature neonates are surviving, rates of neurodevelopmental impairment are not improving. We developed a novel mouse model with clinically relevant exposures to test the hypothesis that systemic maternal inflammation with transient neonatal hyperoxia exposure will induce a phenotype similar to diffuse periventricular leukomalacia (PVL) like that observed in premature human infants. MAIN METHODS: Timed-pregnant C3H/HeN mice received intraperitoneal injections of lipopolysaccharide (LPS) or saline on embryonic day 16. Newborn pups were placed in room air (RA) or 85% oxygen (O2) for 14 days, followed by 14 days in RA recovery. Oligodendroglial and microglial populations were evaluated at 14 and 28 days. KEY FINDINGS: Brain weight to body weight ratios were lower in mice exposed to LPS. Oligodendrocyte numbers were decreased significantly in the cerebral cortex and hippocampus in groups exposed to LPS or LPS/O2 at 14 days, and persisted in the cerebral cortex at 28 days for LPS/O2 mice. At day 14, cleaved caspase 3 was increased and numbers of microglia were elevated in the cerebral cortex and hippocampus of LPS/O2 animals. SIGNIFICANCE: These data indicate that combining systemic maternal LPS and neonatal hyperoxic exposure impairs myelination, and suggests that this novel mouse model may represent a subtle, diffuse form of periventricular white matter injury that could provide a clinically relevant platform for further study of perinatal brain injury.


Assuntos
Encéfalo/citologia , Inflamação/complicações , Oligodendroglia/fisiologia , Complicações na Gravidez/fisiopatologia , Animais , Animais Recém-Nascidos/fisiologia , Encéfalo/fisiopatologia , Caspase 3/metabolismo , Contagem de Células , Córtex Cerebral/citologia , Córtex Cerebral/fisiopatologia , Feminino , Hipocampo/citologia , Hipocampo/fisiopatologia , Hiperóxia/complicações , Hiperóxia/fisiopatologia , Inflamação/fisiopatologia , Lipopolissacarídeos/farmacologia , Camundongos , Microglia/citologia , Microglia/fisiologia , Oligodendroglia/citologia , Gravidez
15.
Am J Physiol Regul Integr Comp Physiol ; 303(3): R279-90, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22718803

RESUMO

Maternally derived inflammatory mediators, such as IL-6 and IL-8, contribute to preterm delivery, low birth weight, and respiratory insufficiency, which are routinely treated with oxygen. Premature infants are at risk for developing adult-onset cardiac, metabolic, and pulmonary diseases. Long-term pulmonary consequences of perinatal inflammation are unclear. We tested the hypothesis that a hostile perinatal environment induces profibrotic pathways resulting in pulmonary fibrosis, including persistently altered lung structure and function. Pregnant C3H/HeN mice injected with LPS or saline on embryonic day 16. Offspring were placed in room air (RA) or 85% O(2) for 14 days and then returned to RA. Pulmonary function tests, microCTs, molecular and histological analyses were performed between embryonic day 18 and 8 wk. Alveolarization was most compromised in LPS/O(2)-exposed offspring. Collagen staining and protein levels were increased, and static compliance was decreased only in LPS/O(2)-exposed mice. Three-dimensional microCT reconstruction and quantification revealed increased tissue densities only in LPS/O(2) mice. Diffuse interstitial fibrosis was associated with decreased micro-RNA-29, increased transforming growth factor-ß expression, and phosphorylation of Smad2 during embryonic or early fetal lung development. Systemic maternal LPS administration in combination with neonatal hyperoxic exposure induces activation of profibrotic pathways, impaired alveolarization, and diminished lung function that are associated with prenatal and postnatal suppression of miR-29 expression.


Assuntos
Hiperóxia/fisiopatologia , Inflamação/fisiopatologia , Pulmão/patologia , Pulmão/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Animais , Colágeno/metabolismo , Feminino , Fibrose , Inflamação/induzido quimicamente , Lipopolissacarídeos/efeitos adversos , Pulmão/embriologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , MicroRNAs/metabolismo , Modelos Animais , Gravidez , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/metabolismo
16.
Exp Lung Res ; 37(3): 155-61, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21128861

RESUMO

The aims of this study were to test the hypothesis that mice expressing mitochondrially targeted human glutathione reductase (GR) driven by a surfactant protein C promoter ((spc-mt)hGR) are functionally riboflavin deficient and that this deficiency exacerbates hyperoxic lung injury. The authors further hypothesized that dietary supplementation with riboflavin (FADH) will improve the bioactivity of GR, thus enhancing resistance to hyperoxic lung injury. Transgenic (mt-spc)hGR mice and their nontransgenic littermates were fed control or riboflavin-supplemented diets upon weaning. At 6 weeks of age the mice were exposed to either room air (RA) or >95% O(2) for up to 84 hours. GR activities (with and without exogenous FADH) and GR protein levels were measured in lung tissue homogenates. Glutathione (GSH) and glutathione disulfide (GSSG) concentrations were assayed to identify changes in GR activity in vivo. Lung injury was assessed by right lung to body weight ratios and bronchoalveolar lavage protein concentrations. The data showed that enhanced GR activity in the mitochondria of lung type II cells does not protect adult mice from hyperoxic lung injury. Furthermore, the addition of riboflavin to the diets of (spc-mt)hGR mice neither enhances GR activities nor offers protection from hyperoxic lung injury. The results indicated that modulation of mitochondrial GR activity in lung type II cells is not an effective therapy to minimize hyperoxic lung injury.


Assuntos
Glutationa Redutase/metabolismo , Hiperóxia/prevenção & controle , Lesão Pulmonar/prevenção & controle , Riboflavina/administração & dosagem , Animais , Glutationa/metabolismo , Glutationa Redutase/genética , Humanos , Hiperóxia/complicações , Hiperóxia/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Lesão Pulmonar/complicações , Lesão Pulmonar/metabolismo , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Proteína C Associada a Surfactante Pulmonar/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Deficiência de Riboflavina/complicações , Deficiência de Riboflavina/tratamento farmacológico
17.
J Appl Physiol (1985) ; 108(5): 1347-56, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20223995

RESUMO

Systemic maternal inflammation contributes to preterm birth and is associated with development of bronchopulmonary dysplasia (BPD). Infants with BPD exhibit decreased alveolarization, diffuse interstitial fibrosis with thickened alveolar septa, and impaired pulmonary function. We tested the hypothesis that systemic prenatal LPS administration to pregnant mice followed by postnatal hyperoxia exposure is associated with prolonged alterations in pulmonary structure and function after return to room air (RA) that are more severe than hyperoxia exposure alone. Timed-pregnant C3H/HeN mice were dosed with LPS (80 microg/kg) or saline on gestation day 16. Newborn pups were exposed to RA or 85% O2 for 14 days and then to RA for an additional 14 days. Data were collected and analyzed on postnatal days 14 and 28. The combination of prenatal LPS and postnatal hyperoxia exposure generated a phenotype with more inflammation (measured as no. of macrophages per high-power field) than either insult alone at day 28. The combined exposures were associated with a diffuse fibrotic response [measured as hydroxyproline content (microg)] but did not induce a more severe developmental arrest than hyperoxia alone. Pulmonary function tests indicated that hyperoxia, independent of maternal exposure, induced compliance decreases on day 14 that did not persist after RA recovery. Either treatment alone or combined induced an increase in resistance on day 14, but the increase persisted on day 28 only in pups receiving the combined treatment. In conclusion, the combination of systemic maternal inflammation and neonatal hyperoxia induced a prolonged phenotype of arrested alveolarization, diffuse fibrosis, and impaired lung mechanics that mimics human BPD. This new model should be useful in designing studies of specific mechanisms and interventions that could ultimately be utilized to define therapies to prevent BPD in premature infants.


Assuntos
Displasia Broncopulmonar/fisiopatologia , Hiperóxia/fisiopatologia , Inflamação/fisiopatologia , Alvéolos Pulmonares/fisiopatologia , Fatores Etários , Resistência das Vias Respiratórias , Animais , Animais Recém-Nascidos , Displasia Broncopulmonar/imunologia , Displasia Broncopulmonar/patologia , Modelos Animais de Doenças , Feminino , Fibrose , Idade Gestacional , Humanos , Hiperóxia/imunologia , Hiperóxia/patologia , Recém-Nascido , Inflamação/imunologia , Inflamação/patologia , Lipopolissacarídeos , Complacência Pulmonar , Macrófagos Alveolares/imunologia , Camundongos , Camundongos Endogâmicos C3H , Neutrófilos/imunologia , Tamanho do Órgão , Fenótipo , Gravidez , Alvéolos Pulmonares/crescimento & desenvolvimento , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/patologia
18.
Am J Respir Cell Mol Biol ; 39(6): 683-8, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18566333

RESUMO

Exposure of the lung epithelium to reactive oxygen species without adequate antioxidant defenses leads to airway inflammation, and may contribute to lung injury. Glutathione peroxidase catalyzes the reduction of peroxides by oxidation of glutathione (GSH) to glutathione disulfide (GSSG), which can in turn be reduced by glutathione reductase (GR). Increased levels of GSSG have been shown to correlate negatively with outcome after oxidant exposure, and increased GR activity has been protective against hyperoxia in lung epithelial cells in vitro. We tested the hypothesis that increased GR expression targeted to type II alveolar epithelial cells would improve outcome in hyperoxia-induced lung injury. Human GR with a mitochondrial targeting sequence was targeted to mouse type II cells using the SPC promoter. Two transgenic lines were identified, with Line 2 having higher lung GR activities than Line 1. Both transgenic lines had lower lung GSSG levels and higher GSH/GSSG ratios than wild-type. Six-week-old wild-type and transgenic mice were exposed to greater than 95% O2 or room air (RA) for 84 hours. After exposure, Line 2 mice had higher right lung/body weight ratios and lavage protein concentrations than wild-type mice, and both lines 1 and 2 had lower GSSG levels than wild-type mice. These findings suggest that GSSG accumulation in the lung may not play a significant role in the development of hyperoxic lung injury, or that compensatory responses to unregulated GR expression render animals more susceptible to hyperoxic lung injury.


Assuntos
Glutationa Redutase/metabolismo , Hiperóxia/complicações , Hiperóxia/enzimologia , Pneumopatias/complicações , Pneumopatias/enzimologia , Alvéolos Pulmonares/enzimologia , Ar , Animais , Glutationa/metabolismo , Humanos , Hiperóxia/patologia , Pulmão/enzimologia , Pulmão/patologia , Pneumopatias/patologia , Camundongos , Oxigênio , Sinais Direcionadores de Proteínas , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia , Proteína C Associada a Surfactante Pulmonar/metabolismo , Riboflavina/farmacologia , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...