Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochim Biophys Acta ; 1475(3): 337-45, 2000 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-10913834

RESUMO

Hox proteins control structural morphogenesis, pattern formation and cell fate in the developing embryo. To determine if Hoxb-5 participates in patterning of early airway branching during lung morphogenesis, gestational day 11.5 embryonic lung cultures were treated with retinoic acid (RA) to up-regulate and antisense oligonucleotides to down-regulate Hoxb-5 protein expression. RA (10(-6) M) and Hoxb-5 antisense oligonucleotide (20 microM) treatment each significantly decreased branching morphogenesis (P<0. 001), but the morphology of branching under these conditions was very different. RA-treated lungs had elongated primary branches but decreased further branching with increased Hoxb-5 immunostaining in subepithelial regions underlying these elongated airways. Western blots confirmed that Hoxb-5 protein was increased by 189+/-20% (mean+/-S.E.M., P<0.05) in RA-treated lungs compared to controls. In contrast, lungs treated with Hoxb-5 antisense oligos plus RA had foreshortened primary branches with rudimentary distal clefts resulting in decreased numbers of primary and subsequent branches. Immunohistochemistry confirmed that Hoxb-5 antisense oligos inhibited Hoxb-5 protein expression even in the presence of RA. We conclude that regional and quantitative changes in Hoxb-5 protein expression influence morphogenesis of the first airway divisions from the mainstem bronchi. RA-induced alterations in branching are mediated in part through regulated Hoxb-5 expression.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Pulmão/embriologia , Animais , Western Blotting , Feminino , Idade Gestacional , Proteínas de Homeodomínio/análise , Imuno-Histoquímica , Pulmão/efeitos dos fármacos , Camundongos , Oligonucleotídeos Antissenso/farmacologia , Técnicas de Cultura de Órgãos , Alvéolos Pulmonares/embriologia , Tretinoína/farmacologia
2.
Pediatr Res ; 47(4 Pt 1): 481-91, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10759155

RESUMO

Early gestation lung development is characterized by branching morphogenesis of the airways and basic lung structure formation. Androgens delay late-gestation lung development if the androgen exposure begins in early gestation. We hypothesized that there would be effects of early gestation androgens on lung development. Embryonic mouse lungs (d 11.5) were cultured with dihydrotestosterone (DHT), DHT plus flutamide, or with nothing as controls. Branching morphogenesis was significantly increased after 24, 48, and 72 h of culture. This effect was blocked by simultaneous flutamide treatment. Fetal sex did not influence the DHT response. DHT increased cell proliferation as measured by [3H]thymidine incorporation into DNA. Autoradiography showed prominent [3H]thymidine labeling of epithelia and mesenchyme in regions of new bud formation. DHT treatment significantly increased the thymidine-labeling index of fibroblasts and airway epithelial cells. Programmed cell death, which is found in developing organs in association with cell proliferation during structure formation and tissue remodeling, was studied using terminal deoxyribonucleotidyl transferase-mediated dUTP-digoxigenin nick end labeling assay. In control lungs, programmed cell death occurred in the peripheral mesenchyme surrounding newly forming buds and underlying airway branch points. DHT treatment increased programmed cell death in association with increased branching morphogenesis. Evaluation of near-adjacent sections (control and DHT-treated lungs) showed that apoptotic mesenchymal cells were flanked by [3H]thymidine-labeled fibroblasts and epithelial cells, suggesting a coordination of these processes in the progression of branching morphogenesis. We conclude that androgen enhances the process of early lung morphogenesis by increasing cell proliferation and programmed cell death and by promoting the structural progression of branching morphogenesis.


Assuntos
Apoptose/efeitos dos fármacos , Di-Hidrotestosterona/farmacologia , Pulmão/efeitos dos fármacos , Morfogênese/efeitos dos fármacos , Animais , Autorradiografia , Sequência de Bases , Primers do DNA , Marcação In Situ das Extremidades Cortadas , Técnicas In Vitro , Pulmão/citologia , Pulmão/embriologia , Camundongos , Timidina/metabolismo
3.
Hum Mol Genet ; 7(4): 597-608, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9499412

RESUMO

Human chromosome 11p15.5 and distal mouse chromosome 7 include a megabase-scale chromosomal domain with multiple genes subject to parental imprinting. Here we describe mouse and human versions of a novel imprinted gene, IMPT1 , which lies between IPL and p57 KIP2 and which encodes a predicted multi-membrane-spanning protein similar to bacterial and eukaryotic polyspecific metabolite transporter and multi-drug resistance pumps. Mouse Impt1 and human IMPT1 mRNAs are highly expressed in tissues with metabolite transport functions, including liver, kidney, intestine, extra-embryonic membranes and placenta, and there is strongly preferential expression of the maternal allele in various mouse tissues at fetal stages. In post-natal tissues there is persistent expression, but the allelic bias attenuates. An allelic expression bias is also observed in human fetal and post-natal tissues, but there is significant interindividual variation and rare somatic allele switching. The fact that Impt1 is relatively repressed on the paternal allele, together with data from other imprinted genes, allows a statistical conclusion that the primary effect of human chromosome 11p15.5/mouse distal chromosome 7 imprinting is domain-wide relative repression of genes on the paternal homolog. Dosage regulation of the metabolite transporter gene(s) by imprinting might regulate placental and fetal growth.


Assuntos
Genes MDR/genética , Impressão Genômica , Proteínas de Membrana/genética , Camundongos/genética , Proteínas de Transporte de Cátions Orgânicos , Adulto , Alelos , Sequência de Aminoácidos , Animais , Proteínas de Transporte , Criança , Cromossomos Humanos Par 11 , Humanos , Recém-Nascido , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Transportador 1 de Cátions Orgânicos , Placenta/metabolismo , Polimorfismo Genético , Distribuição Tecidual
4.
Am J Physiol ; 274(4): L610-20, 1998 04.
Artigo em Inglês | MEDLINE | ID: mdl-9575880

RESUMO

Studies on lung morphogenesis have indicated a role of homeobox (Hox) genes in the regulation of lung development. In the present study, we attempted to modulate the synthesis of Hoxb5 protein in cultured murine fetal lungs after mechanical or chemical stimuli. Murine fetuses at gestational day 14 (GD14) were removed from pregnant CD-1 mice, and lungs were excised and cultured for 7 days in BGJb media. The experimental groups were 1) untreated, unligated; 2) tracheal ligation; 3) supplemented media with either epidermal growth factor (EGF; 10 ng/ml), transforming growth factor (TGF)-beta 1 (2 ng/ml), dexamethasone (10 nM), EGF + TGF-beta 1, or EGF + TGF-beta 1 + dexamethasone. After 3 or 7 days, the cultured lungs were compared with in vivo lungs. Immunoblotting signals at 3 days in culture were stronger than those at 7 days. Western blot analyses showed that ligation, EGF, TGF-beta 1, and EGF + TGF-beta 1 downregulated Hoxb5 protein to approximately 20-70% of Hoxb5 protein levels in unligated, untreated cultured lungs. Furthermore, dexamethasone alone or in combination with EGF and TGF-beta 1 downregulated Hoxb5 protein by > 90% (P < 0.05) signal strength, similar to that seen in GD19 or in neonatal lungs. Immunostaining showed that Hoxb5 protein was expressed strongly in the lung mesenchyme at early stages in gestation. However, by GD19 and in neonates, it was present only in specific epithelial cells. A persistent level of Hoxb5 protein in the mesenchyme after EGF or TGF-beta 1 treatments or tracheal ligation was noted. Hoxb5 protein was significantly downregulated by EGF + TGF-beta 1, and it was least in lungs after dexamethasone or EGF + TGF-beta 1 + dexamethasone treatment. The decrease in Hoxb5 protein was significant only in the groups with dexamethasone added to the media. Thus immunostaining results parallel those of immunoblotting. The degree of Hoxb5 downregulation by dexamethasone or EGF + TGF-beta 1 + dexamethasone was similar to that seen in vivo in very late gestation, which correlated to the advancing structural development of the lung.


Assuntos
Dexametasona/farmacologia , Glucocorticoides/farmacologia , Substâncias de Crescimento/farmacologia , Proteínas de Homeodomínio/metabolismo , Pulmão/metabolismo , Animais , Técnicas de Cultura , Feto/metabolismo , Immunoblotting , Imuno-Histoquímica , Pulmão/embriologia , Camundongos/embriologia , Camundongos Endogâmicos , Fatores de Tempo
5.
Am J Obstet Gynecol ; 178(2): 242-6, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9500481

RESUMO

OBJECTIVE: Alterations in maternal plasma arginine concentration accompany normal pregnancy. Nitric oxide is synthesized from L-arginine and influences fetal growth. We hypothesized that L-arginine would influence fetal growth and hypoxia-induced uricemia in a maternal hypoxia-induced fetal growth restriction model. STUDY DESIGN: Fetal growth on day 21 of gestation was assessed in timed pregnant Wistar rats with or without exposure to maternal hypobaric hypoxia. Animals exposed to hypoxia received either no supplement or supplementation of drinking water with 0.2% L-arginine, 2% L-arginine, or 2% glycine. On day 21 of gestation, fetuses were delivered by hysterotomy and fetal and placental weights were obtained. Maternal and fetal plasma were assayed for uric acid as an index of tissue hypoxia. Xanthine oxidase and xanthine dehydrogenase, precursors of uric acid and reactive oxygen species, were assayed in maternal tissue. Results were analyzed by analysis of variance with correction for multiple comparisons. RESULTS: Exposure of rats on normal diets to hypoxia resulted in a 30% reduction in fetal weights. L-Arginine, 2% or 0.2%, prevented the reduction in fetal weight (p < 0.0001). Isocaloric and isonitrogenous supplementation with glycine did not influence hypoxia-induced fetal growth restriction. CONCLUSION: L-Arginine, but not glycine, ameliorates maternal hypoxia-induced fetal growth restriction in the rat.


Assuntos
Arginina/administração & dosagem , Dieta , Retardo do Crescimento Fetal/prevenção & controle , Animais , Pressão Atmosférica , Desenvolvimento Embrionário e Fetal/efeitos dos fármacos , Endotelinas/sangue , Feminino , Retardo do Crescimento Fetal/etiologia , Glicina/administração & dosagem , Hipóxia , Óxido Nítrico/metabolismo , Gravidez , Ratos , Ratos Wistar , Ácido Úrico/sangue , Xantina Desidrogenase/metabolismo , Xantina Oxidase/metabolismo
6.
Biochem Mol Med ; 60(1): 38-48, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9066980

RESUMO

embryonic lung cultures were exposed to either EGF (10 ng/ml) or TGF beta 1 (2 ng/ml) for 72 h, and branching morphogenesis, cell proliferation, and epithelial differentiation (the expression of DSPC synthesis and of surfactant protein C (SP-C) mRNA) were studied. EGF treatment stimulated branching morphogenesis (measured as the number of terminal left lung buds), epithelial differentiation, and cell proliferation. Branching morphogenesis was increased compared to controls after 48 h of culture by 47% and after 72 h by 34% (P < 0.0005). Choline incorporation into DSPC was stimulated by 343% (P = 0.05). SP-C expression was increased sixfold. Thymidine incorporation was stimulated by 49% (P < 0.05). The effects of EGF on thymidine labeling were distributed among epithelial cells of the airway walls and of the branching tips, and also the mesenchyme (P < 0.01 for each area compared to controls). In contrast, TGF beta 1 did not alter the number of terminal left lung buds, inhibited choline incorporation into DSPC by 35% (P < 0.05), and had no effect on thymidine incorporation (87% of control). There was increased thymidine labeling at the branching tips (P < 0.01), while other areas were not different from controls. We conclude that both EGF and TGF beta 1 affect the development of branching morphogenesis and of epithelial differentiation in the embryonic lung.


Assuntos
Fator de Crescimento Epidérmico/fisiologia , Pulmão/embriologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Sequência de Bases , Diferenciação Celular , Divisão Celular , Primers do DNA , Epitélio , Feminino , Técnicas In Vitro , Camundongos , Índice Mitótico , Dados de Sequência Molecular , Morfogênese , Proteolipídeos/metabolismo , Surfactantes Pulmonares/metabolismo
7.
Histochem Cell Biol ; 108(6): 495-504, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9450632

RESUMO

Hoxb-5 is one of the few homeobox genes strongly expressed in the developing mouse lung. To explore the hypothesis that Hoxb-5 acts to regulate epithelial cell fate and branching morphogenesis in the developing lung, we studied the temporal, spatial, and cell-specific expression of Hoxb-5 from gestational day (d) 13.5 to postnatal day (P) 2. Immunocytochemistry demonstrated regional localization of Hoxb-5 protein to developing conducting airways and surrounding mesenchyme. The cellular expression pattern changed from diffusely positive nuclei of mesenchymal cells on d13.5 to become more localized to nuclei of subepithelial fibroblasts and some adjacent columnar and cuboidal epithelial cells on d14.5. After d14.5, Hoxb-5 protein expression continued to decrease in mesenchymal cells distal from developing airways, but persisted in fibroblasts underlying conducting airways. Hoxb-5 protein expression persisted in nuclei of columnar and cuboidal epithelial cells on d16.5 and d17.5, with expression in low cuboidal epithelial cells as well from d17.5 to P2. Western blot analysis showed temporal and quantitative changes in Hoxb-5 protein expression with peak expression on d14.5-15.5. We conclude that Hoxb-5 protein is developmentally regulated in a temporal, spatial, and cell-specific manner throughout the pseudoglandular, canalicular, and terminal saccular periods of lung development in the mouse. This localization and expression pattern suggests that Hoxb-5 may influence branching morphogenesis, cell-cell communication, cell fate, and differentiation of conducting airway epithelia.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Genes Homeobox , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/fisiologia , Pulmão/embriologia , Pulmão/metabolismo , Animais , Western Blotting , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Imuno-Histoquímica , Camundongos , Gravidez
8.
Am J Physiol ; 271(5 Pt 1): L869-74, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8944732

RESUMO

We have previously shown that nitric oxide (NO) donors, such as nitrosoglutathione, inhibit endothelial cell (EC) xanthine dehydrogenase (XD)/xanthine oxidase (XO) activity. The purpose of this study was to assess whether endothelial-derived NO plays any role in the regulation of intracellular XD/XO. We exposed rat pulmonary microvascular EC to L-arginine (precursor of NO) or inhibitors of nitric oxide synthase (NOS), i.e., NG-nitro-L-arginine methyl esther (L-NAME) and NG-nitro-L-arginine, in conditions of normoxia, hypoxia, and hypoxia followed by reoxygenation. Hypoxia alone caused a 1.9- and a 6.6-fold increase in XO and a 5-fold increase in XO + XD activities after 24 and 48 h of exposure, respectively. The combination of hypoxia and L-NAME (300 microM) treatment amounted at 48 h to a 10- and 7.5-fold increase in XO and XO + XD activities, respectively, compared with normoxic untreated cells. L-NAME also prevented the decline in XD/XO activity that occurred in untreated EC after hypoxia-reoxygenation. On the other hand, treatment with L-arginine caused a dose-dependent decrease in XD/XO activity in hypoxic EC compared with cells provided with L-arginine-free medium. In separate experiments, we assessed the role of L-arginine supplementation on the in vivo regulation of lung XD/XO by exposing male adult Sprague-Dawley rats for a period of 5 days to a hypoxic hypobaric atmosphere (0.5 atm). Exposure to hypoxia produced a significant increase in lung tissue XO activity and an increase in the ratio of XO to XD. L-Arginine supplementation in the drinking water prevented the increase in lung XO and the XO-to-XD ratio in hypoxic rats and caused a significant decrease in XO and XD in rats exposed to normoxia. In conclusion, this study suggests that endogenous NO has a significant role in the regulation of XD/XO both in vitro and in vivo. By inhibiting XD/XO activity, NO may have a modulating effect in conditions of hypoxia and hypoxia-reoxygenation, where this enzyme is thought to be important.


Assuntos
Endotélio Vascular/enzimologia , Circulação Pulmonar , Xantina Desidrogenase/metabolismo , Xantina Oxidase/metabolismo , Animais , Arginina/farmacologia , Hipóxia Celular , Células Cultivadas , Endotélio Vascular/efeitos dos fármacos , Cinética , Masculino , Microcirculação , NG-Nitroarginina Metil Éster/farmacologia , Oxigênio/farmacologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
9.
Biol Neonate ; 66(2-3): 65-70, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-7993949

RESUMO

Persistent pulmonary hypertension of the newborn (PPHN) is a life-threatening disorder of neonates manifested by vasoconstriction of the pulmonary arteries. Recently, the gas nitric oxide (NO) has been used with some success in the management of infants with PPHN. Exogenous administration of NO selectively dilates the pulmonary vascular bed. NO is naturally synthesized in the body from the amino acid L-arginine. Here we report our findings that infants with PPHN are deficient in arginine and achieve normal or elevated plasma arginine concentrations with intravenous hyperalimentation. We prospectively identified and studied 10 infants with PPHN who were not receiving protein or amino acids for at least 24 h and compared their plasma arginine concentrations to 8 control infants without PPHN given similar nutrition. Plasma arginine concentrations were 32 +/- 14 and 52 +/- 20 mumol/l in infants with PPHN and control infants, respectively (p = 0.02). There were no other statistically significant differences in plasma amino acid concentration for any of the 22 other amino acids determined. Infants with PPHN who were subsequently treated with amino acid infusions had plasma arginine concentrations of 115 +/- 48 mumol/l (mean of ten determinations at 86 +/- 27 h after initiation of intravenous amino acids in five PPHN infants).


Assuntos
Arginina/deficiência , Síndrome da Persistência do Padrão de Circulação Fetal/complicações , Aminoácidos/administração & dosagem , Arginina/sangue , Feminino , Humanos , Recém-Nascido , Masculino , Óxido Nítrico/metabolismo , Nutrição Parenteral , Síndrome da Persistência do Padrão de Circulação Fetal/diagnóstico , Síndrome da Persistência do Padrão de Circulação Fetal/terapia , Estudos Prospectivos
10.
Prenat Diagn ; 13(12): 1139-41, 1993 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8177833

RESUMO

A case of fetal pleural effusion in a fetus affected with Duchenne muscular dystrophy (DMD) is reported. This case is discussed in the context of the previous observation of frequent stillbirths among male fetuses in DMD families.


Assuntos
Distrofias Musculares/complicações , Derrame Pleural/complicações , Poli-Hidrâmnios/complicações , Ultrassonografia Pré-Natal , Adulto , Feminino , Humanos , Masculino , Distrofias Musculares/diagnóstico por imagem , Distrofias Musculares/genética , Derrame Pleural/diagnóstico por imagem , Poli-Hidrâmnios/diagnóstico por imagem , Gravidez
12.
J Cell Physiol ; 138(3): 493-502, 1989 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-2494195

RESUMO

The c-fos gene, the cellular homologue of the transforming gene of the FBJ osteosarcoma virus, v-fos, is strongly induced in quiescent BALB/c 3T3 cells by growth factors and in other cell types by a wide variety of transmembrane signalling agents. c-fos is a member of a family of structurally related proteins which includes the fos-related antigens (fra). We have studied the dynamic state of the c-fos protein with an antibody prepared by immunizing rabbits with a plasmid-encoded fos fusion protein. In serum-stimulated BALB/c 3T3 cells, the antibody recognizes a nuclear antigen which resolves on SDS-PAGE as a 60-68-kD group of bands corresponding to c-fos, a doublet at 44-45-kD corresponding to the noncovalently associated p39 protein, as well as an approximately 50-kD band corresponding to a fra. We show that although c-fos protein synthesis is only transiently induced by serum, the c-fos protein persists within the cell after its synthesis has ceased, and it decays with a half-life of 2 hours. Significantly, newly synthesized p39 continues to appear in the immune-precipitated complex even at times when c-fos is no longer synthesized. These kinetics indicate that even following shutoff of c-fos protein synthesis, p39 is newly synthesized and can complex with c-fos protein or a fos-related antigen. During this time, c-fos also undergoes an extensive posttranslational modification. The modification is partially reversed by phosphatase treatment, which implicates protein phosphorylation. Together these results suggest that both interaction with p39 and phosphorylation may progressively modify the properties of c-fos and/or the fos-related antigens over a period of 4-8 hours following the shutoff of fos synthesis. We discuss the implications of the dynamic state of c-fos and fra protein interactions for the function of these proteins.


Assuntos
Ciclo Celular , Proteínas Proto-Oncogênicas/metabolismo , Animais , Especificidade de Anticorpos , Western Blotting , Linhagem Celular , Regulação da Expressão Gênica , Técnicas Imunológicas , Substâncias Macromoleculares , Camundongos , Fosfoproteínas/fisiologia , Testes de Precipitina , Proteínas Proto-Oncogênicas c-fos , Proteínas Recombinantes de Fusão/imunologia
13.
Mol Cell Biol ; 8(4): 1670-6, 1988 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-2454395

RESUMO

Transcription of the protooncogene c-fos is increased greater than 10-fold within minutes of treatment of fibroblasts with serum or purified growth factors. Recent experiments with mouse 3T3 cell lines containing inducible fos antisense RNA constructs have shown that induced fos antisense RNA transcripts cause either a marked inhibition of growth in continuously proliferating cells or, conversely, a minimal effect except during the transition from a quiescent (G0) state into the cell cycle. Since intracellular production of large amounts of antisense RNA does not completely block gene expression, we microinjected affinity-purified antibodies raised against fos to determine whether and when during the cell cycle c-fos expression was required for cell proliferation. Using this independent method, we found that microinjected fos antibodies efficiently blocked serum-stimulated DNA synthesis when injected up to 6 to 8 h after serum stimulation of quiescent REF-52 fibroblasts. Furthermore, when fos antibodies were injected into asynchronously growing cells, a consistently greater number of cells was prevented from synthesizing DNA than when cells were injected with nonspecific immunoglobulins. Thus, whereas the activity of c-fos may be necessary for transition of fibroblasts from G0 to G1 of the cell cycle, its function is also required during the early G1 portion of the cell cycle to allow subsequent DNA synthesis.


Assuntos
Anticorpos , Replicação do DNA , DNA/imunologia , Proto-Oncogenes , Transcrição Gênica , Animais , Anticorpos/administração & dosagem , Ciclo Celular , Linhagem Celular , Células Cultivadas , DNA/genética , Fibroblastos/citologia , Cinética , Microinjeções , RNA/genética , RNA Antissenso , RNA Mensageiro/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...