Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Biomark ; 33(4): 479-488, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35491772

RESUMO

NASA's Jet Propulsion Laboratory (JPL) is advancing research capabilities for data science with two of the National Cancer Institute's major research programs, the Early Detection Research Network (EDRN) and the Molecular and Cellular Characterization of Screen-Detected Lesions (MCL), by enabling data-driven discovery for cancer biomarker research. The research team pioneered a national data science ecosystem for cancer biomarker research to capture, process, manage, share, and analyze data across multiple research centers. By collaborating on software and data-driven methods developed for space and earth science research, the biomarker research community is heavily leveraging similar capabilities to support the data and computational demands to analyze research data. This includes linking diverse data from clinical phenotypes to imaging to genomics. The data science infrastructure captures and links data from over 1600 annotations of cancer biomarkers to terabytes of analysis results on the cloud in a biomarker data commons known as "LabCAS". As the data increases in size, it is critical that automated approaches be developed to "plug" laboratories and instruments into a data science infrastructure to systematically capture and analyze data directly. This includes the application of artificial intelligence and machine learning to automate annotation and scale science analysis.


Assuntos
Inteligência Artificial , Ciência de Dados , Biomarcadores Tumorais , Ecossistema , Humanos , Software
2.
Biol Lett ; 7(2): 285-7, 2011 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-20880860

RESUMO

Heterochrony, differences in the timing of developmental events between descendent species and their ancestors, is a pervasive evolutionary pattern. However, the origins of such timing changes are still not resolved. Here we show, using sequence analysis, that exposure to predator cues altered the timing of onset of several developmental events in embryos of two closely related gastropod species: Radix balthica and Radix auricularia. These timing alterations were limited to certain events and were species-specific. Compared with controls, over half (62%) of exposed R. auricularia embryos had a later onset of body flexing and an earlier occurrence of the eyes and the heart; in R. balthica, 67 per cent of exposed embryos showed a later occurrence of mantle muscle flexing and an earlier attachment to, and crawling on, the egg capsule wall. The resultant developmental sequences in treated embryos converged, and were more similar to one another than were the sequences of the controls for both species. We conclude that biotic agents can elicit altered event timing in developing gastropod embryos. These changes were species-specific, but did not occur in all individuals. Such developmental plasticity in the timing of developmental events could be an important step in generating interspecific heterochrony.


Assuntos
Sinais (Psicologia) , Desenvolvimento Embrionário , Lymnaea/embriologia , Comportamento Predatório , Animais , Olho/embriologia , Peixes/fisiologia , Coração/embriologia , Lymnaea/crescimento & desenvolvimento , Especificidade da Espécie , Fatores de Tempo
3.
Eur Phys J E Soft Matter ; 30(2): 117-21, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19777278

RESUMO

The fundamental study of the adhesion of cells to each other or to a substrate is a key research topic in cellular biophysics because cell adhesion is important to many biological processes. We report on the adhesion of a model cell, a liposome, and a living HeLa cell to a substrate measured with a novel experimental technique. The cells are held at the end of a micropipette mounted on a micromanipulator and brought into contact with a surface. The adhesion energy and membrane tension are measured directly using the deflection of the micropipette when binding or unbinding the cell from the substrate. Since the force applied on the cells is known throughout the experiment, the technique presented enables the measurement of dynamics such as changes in the adhesion, elasticity, and membrane tension with time.


Assuntos
Biofísica/métodos , Membrana Celular/fisiologia , Vesículas Transportadoras/fisiologia , Adesão Celular , Células HeLa , Humanos , Nanotecnologia/métodos , Estresse Mecânico , Tensão Superficial
5.
Dev Biol ; 232(2): 351-61, 2001 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11401397

RESUMO

Retinoic acid (RA), a potent teratogen, produces a characteristic set of embryonic cardiovascular malformations similar to those observed in neural crest ablated avians. While the effects of RA on neural crest are well described, the molecular mechanism(s) of RA action on these cells is less clear. The present study examines the relationship between RA and mitogen-activated protein kinase signaling in neural crest cells and demonstrates that c-Jun N-terminal kinase (JNK) activation is severely repressed by RA. RA suppressed migration and proliferation of primary cultures of mouse neural crest cells treated in vitro as well as from animals treated in vivo. On Western blots, JNK activation/phosphorylation in neural crest cultures was reduced, while neither extracellular signal-regulated kinase (ERK) nor p38 pathways were affected. Both the dose-dependent stimulation of neural crest outgrowth and JNK phosphorylation by platelet-derived growth factor AA, which promotes outgrowth but not proliferation of neural crest cultures, were completely abrogated by RA. To establish the relevance of the JNK signaling pathway to cardiac neural crest migration, dominant negative adenoviral constructs were used to inhibit upstream activation of JNK or c-Jun downstream responses. Both adenoviral constructs markedly reduced neural crest cell outgrowth, while a dominant negative inhibitor of the p38 pathway had no effect. These data demonstrate that the JNK signaling pathway and c-Jun activation are critical for cardiac neural crest outgrowth and are potential targets for the action of RA.


Assuntos
Coração Fetal/efeitos dos fármacos , Coração Fetal/inervação , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Crista Neural/efeitos dos fármacos , Tretinoína/toxicidade , Animais , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Coração Fetal/enzimologia , Proteínas Quinases JNK Ativadas por Mitógeno , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Crista Neural/citologia , Crista Neural/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Transdução de Sinais , Teratogênicos/toxicidade , Fator de Transcrição AP-1/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
6.
Nature ; 410(6828): 539-40, 2001 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-11279483

RESUMO

The Archaeoraptor fossil was announced as a 'missing link' and purported to be possibly the best evidence since Archaeopteryx that birds did, in fact, evolve from certain types of carnivorous dinosaur. It reportedly came from Early Cretaceous beds of China that have produced other spectacular fossils transitional between birds and extinct non-avian dinosaurs. But Archaeoraptor was revealed to be a forgery in which bones of a primitive bird and a non-flying dromaeosaurid dinosaur had been combined. Here we use high-resolution X-ray computed tomography (CT) to determine the nature and extent of the forgery, as well as how it was built, by imaging the fracture pattern and distribution of materials through the entire specimen.


Assuntos
Dinossauros , Fósseis , Fraude , Animais , Evolução Biológica , Aves , China , Tomografia Computadorizada por Raios X
7.
Pediatr Res ; 48(1): 36-42, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10879798

RESUMO

In children, dilated cardiomyopathy is due to a variety of etiologies and usually carries a grave prognosis. The purpose of the present study was to carefully follow the progression of events leading to cardiac dilatation and congestive heart failure in a dilated cardiomyopathy model in neonatal and juvenile mice. These initial steps are often not well characterized. Furthermore, the loss of gap junctions and reduced electrical coupling of cardiomyocytes frequently found in human cardiomyopathies are also observed in these early stages. By 2 wk of age, molecular markers associated with hypertrophy were already altered. Cardiomyocyte hypertrophy, reduced connexin43 expression, and decreased conduction velocity were apparent by 4 wk, before overt cardiac dysfunction (decreased shortening fraction and chamber remodeling) that was not present until 12 wk of age. Our results show that in this model cardiomyopathic changes are present by 2 wk after birth and progress rapidly during the subsequent 2 postnatal weeks. Combined with the observations of other models of heart disease, we suggest that the first 2 wk of postnatal life are absolutely critical for normal cardiac development, and events that perturb homeostasis during this period determine whether the heart will continue to develop normally. These animals exhibit early symptoms of disease including reduced connexin43 and conduction defects before impaired cardiac function and demonstrate for the first time a temporal association between decreased connexin43 levels and the initiation of a contractility deficit that ends in heart failure.


Assuntos
Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/fisiopatologia , Insuficiência Cardíaca/fisiopatologia , Miocárdio/patologia , Cadeias Pesadas de Miosina/genética , Envelhecimento , Animais , Animais Recém-Nascidos , Cardiomiopatia Dilatada/patologia , Conexina 43/análise , Progressão da Doença , Ecocardiografia , Coração/fisiopatologia , Sistema de Condução Cardíaco/fisiopatologia , Insuficiência Cardíaca/patologia , Humanos , Camundongos , Camundongos Transgênicos , Cadeias Pesadas de Miosina/fisiologia
9.
J Clin Invest ; 103(9): 1277-85, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10225971

RESUMO

The Ron/STK receptor tyrosine kinase is a member of the c-Met family of receptors and is activated by hepatocyte growth factor-like protein (HGFL). Ron activation results in a variety of cellular responses in vitro, such as activation of macrophages, proliferation, migration, and invasion, suggesting a broad biologic role in vivo. Nevertheless, HGFL-deficient mice grow to adulthood with few appreciable phenotypic abnormalities. We report here that in striking contrast to the loss of its only known ligand, complete loss of Ron leads to early embryonic death. Embryos that are devoid of Ron (Ron-/-) are viable through the blastocyst stage of development but fail to survive past the peri-implantation period. In situ hybridization analysis demonstrates that Ron is expressed in the trophectoderm at embryonic day (E) 3.5 and is maintained in extraembryonic tissue through E7.5, compatible with an essential function at this stage of development. Hemizygous mice (Ron+/-) grow to adulthood; however, these mice are highly susceptible to endotoxic shock and appear to be compromised in their ability to downregulate nitric oxide production. These results demonstrate a novel role for Ron in early mouse development and suggest that Ron plays a limiting role in the inflammatory response.


Assuntos
Implantação do Embrião , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Sequência de Bases , Primers do DNA , Desenvolvimento Embrionário e Fetal , Feminino , Morte Fetal/genética , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/metabolismo , Hibridização In Situ , Camundongos , Óxido Nítrico/fisiologia , Receptores Proteína Tirosina Quinases/genética , Receptores de Superfície Celular/genética , Choque Séptico/genética , Células-Tronco/metabolismo
10.
Endocrinology ; 140(4): 1826-33, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10098521

RESUMO

PTH-related protein (PTHrP) is produced in vascular smooth muscle, where it is postulated to exert vasorelaxant properties by activation of the PTH/PTHrP type 1 receptor. As a model for studying the actions of locally produced PTHrP in vascular smooth muscle in vivo, we developed transgenic mice that overexpress the PTH/PTHrP receptor (PTHrP-R) in smooth muscle. Oocyte injection with a SMP8-PTHrP-R fusion construct yielded six founder mice. F1 offspring were viable and demonstrated selective overexpression of the SMP8-PTHP-R messenger RNA in smooth muscle-rich tissues. Baseline blood pressure measured in conscious mice by tail sphygmomanometry was significantly lower in the receptor-overexpressing mice than that in controls (117 +/- 4 vs. 133 +/- 3 mm Hg; P < 0.05). In anesthetized animals, iv infusion of PTHrP-(1-34)NH2 caused a significantly greater reduction in blood pressure and total peripheral resistance in transgenic mice than in control animals. Vascular contractility was studied in paired, isometrically mounted aortas from 9-week-old transgenic and wild-type mice. The force of contraction in response to phenlyephrine was not significantly different between transgenic and wild-type mice. However, PTHrP-(1-34) NH2 relaxed aortic vessel preparations from transgenic mice to a greater extent than in controls (77.1 +/- 3% vs. 38.4 +/- 4%; P < 0.001). To determine the impact of overexpression of PTH/PTHrP type 1 receptor and its ligand on the development of the cardiovascular system, double transgenic mice were created by crossing SMP8-PTHrP-R transgenic mice with mice overexpressing PTHrP (SMP8-PTHrP). Double transgenic mice died around day E9 with abnormalities in the developing heart. In conclusion, overexpression of PTH/PTHrP type 1 receptor in vascular smooth muscle of transgenic mice reduces blood pressure, probably through sustained activation of the receptor by endogenous ligand. The cardiovascular defects observed in mice overexpressing both PTHrP and its receptor suggest that PTHrP may play a role in the normal development of the cardiovascular system.


Assuntos
Pressão Sanguínea/fisiologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , Proteínas/farmacologia , Receptores de Hormônios Paratireóideos/genética , Actinas/genética , Animais , Peso Corporal , Expressão Gênica , Hemodinâmica , Camundongos , Camundongos Transgênicos , Contração Muscular/fisiologia , Proteína Relacionada ao Hormônio Paratireóideo , Fragmentos de Peptídeos/farmacologia , Fenilefrina/farmacologia , Receptor Tipo 1 de Hormônio Paratireóideo , Receptores de Hormônios Paratireóideos/fisiologia , Resistência Vascular/efeitos dos fármacos , Vasoconstritores/farmacologia
11.
Science ; 281(5383): 1690-3, 1998 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-9733519

RESUMO

Hypertrophic cardiomyopathy (HCM) is an inherited form of heart disease that affects 1 in 500 individuals. Here it is shown that calcineurin, a calcium-regulated phosphatase, plays a critical role in the pathogenesis of HCM. Administration of the calcineurin inhibitors cyclosporin and FK506 prevented disease in mice that were genetically predisposed to develop HCM as a result of aberrant expression of tropomodulin, myosin light chain-2, or fetal beta-tropomyosin in the heart. Cyclosporin had a similar effect in a rat model of pressure-overload hypertrophy. These results suggest that calcineurin inhibitors merit investigation as potential therapeutics for certain forms of human heart disease.


Assuntos
Inibidores de Calcineurina , Miosinas Cardíacas , Cardiomegalia/prevenção & controle , Cardiomiopatia Dilatada/prevenção & controle , Cardiomiopatia Hipertrófica/prevenção & controle , Ciclosporina/farmacologia , Proteínas dos Microfilamentos , Miocárdio/metabolismo , Tacrolimo/farmacologia , Animais , Calcineurina/metabolismo , Cálcio/metabolismo , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/metabolismo , Cardiomiopatia Hipertrófica/patologia , Proteínas de Transporte/genética , Feminino , Camundongos , Camundongos Transgênicos , Modelos Cardiovasculares , Miocárdio/patologia , Cadeias Leves de Miosina/genética , Cadeias Leves de Miosina/metabolismo , Ratos , Transdução de Sinais , Tropomodulina , Tropomiosina/genética
12.
Proc Natl Acad Sci U S A ; 95(13): 7597-602, 1998 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-9636195

RESUMO

The conversion of prothrombin (FII) to the serine protease, thrombin (FIIa), is a key step in the coagulation cascade because FIIa triggers platelet activation, converts fibrinogen to fibrin, and activates regulatory pathways that both promote and ultimately suppress coagulation. However, several observations suggest that FII may serve a broader physiological role than simply stemming blood loss, including the identification of multiple G protein-coupled, thrombin-activated receptors, and the well-documented mitogenic activity of FIIa in in vitro test systems. To explore in greater detail the physiological roles of FII in vivo, FII-deficient (FII-/-) mice were generated. Inactivation of the FII gene leads to partial embryonic lethality with more than one-half of the FII-/- embryos dying between embryonic days 9.5 and 11.5. Bleeding into the yolk sac cavity and varying degrees of tissue necrosis were observed in many FII-/- embryos within this gestational time frame. However, at least one-quarter of the FII-/- mice survived to term, but ultimately they, too, developed fatal hemorrhagic events and died within a few days of birth. This study directly demonstrates that FII is important in maintaining vascular integrity during development as well as postnatal life.


Assuntos
Morte Fetal , Protrombina/fisiologia , Alelos , Animais , Morte Fetal/genética , Marcação de Genes , Hemorragia/embriologia , Hemorragia/genética , Hipoprotrombinemias/embriologia , Hipoprotrombinemias/genética , Camundongos , Camundongos Knockout , Protrombina/genética , Tempo de Protrombina
13.
Gene ; 198(1-2): 289-96, 1997 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9370294

RESUMO

We describe a simple, rapid technique for simultaneously isolating large numbers of cDNAs encoding secreted proteins. The technique makes use of a facile genetic selection performed in a strain of Saccharomyces cerevisiae deleted for its endogenous invertase gene. A cDNA cloning vector which carries a modified invertase gene lacking its leader sequence is used in conjunction with this strain. Heterologous secreted genes fused appropriately upstream of this defective invertase provide the necessary signals to restore secretion, allowing the yeast to grow on sugars such as sucrose or raffinose. This microbial growth selection facilitates scanning cDNA libraries containing millions of clones, enabling the wholesale identification of novel secreted proteins without the need for specific bioassays. The technique is similar to one previously described (Klein et al. (1996) Proc. Natl. Acad. Sci. USA 93, 7108-7113). We describe results using a cDNA library derived from activated human peripheral blood mononuclear cells (PBMC). Genes identified from this library encoded signal sequences of proteins of diverse structure, function, and cellular location such as cytokines, type 1 and type 2 transmembrane proteins, and proteins found in intracellular organelles. In addition, a number of novel secreted proteins were identified, including a chemokine and a novel G-protein-coupled receptor. Since signal sequences possess features conserved throughout evolution, the procedure can be used to isolate genes encoding secreted proteins from both eukaryotes and prokaryotes.


Assuntos
DNA Complementar/isolamento & purificação , Vetores Genéticos , Sinais Direcionadores de Proteínas , Proteínas/metabolismo , Receptores de Superfície Celular/genética , Sequência de Aminoácidos , Quimiocinas/genética , Glicosídeo Hidrolases/genética , Humanos , Interferon gama/genética , Dados de Sequência Molecular , Saccharomyces cerevisiae/genética , Seleção Genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Homologia de Sequência do Ácido Nucleico , beta-Frutofuranosidase
14.
J Clin Invest ; 100(8): 1958-68, 1997 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-9329959

RESUMO

Retinoids play a critical role in cardiac morphogenesis. To examine the effects of excessive retinoid signaling on myocardial development, transgenic mice that overexpress a constitutively active retinoic acid receptor (RAR) controlled by either the alpha- or beta-myosin heavy chain (MyHC) promoter were generated. Animals carrying the alpha-MyHC-RAR transgene expressed RARs in embryonic atria and in adult atria and ventricles, but developed no signs of either malformations or disease. In contrast, beta-MyHC-RAR animals, where expression was activated in fetal ventricles, developed a dilated cardiomyopathy that varied in severity with transgene copy number. Characteristic postmortem lesions included biventricular chamber dilation and left atrial thrombosis; the incidence and severity of these lesions increased with increasing copy number. Transcript analyses showed that molecular markers of hypertrophy, alpha-skeletal actin, atrial natriuretic factor and beta-MyHC, were upregulated. Cardiac performance of transgenic hearts was evaluated using the isolated perfused working heart model as well as in vivo, by transthoracic M-mode echocardiography. Both analyses showed moderate to severe impairment of left ventricular function and reduced cardiac contractility. Thus, expression of a constitutively active RAR in developing atria and/ or in postnatal ventricles is relatively benign, while ventricular expression during gestation can lead to significant cardiac dysfunction.


Assuntos
Cardiomiopatia Dilatada/genética , Cardiopatias Congênitas/genética , Insuficiência Cardíaca/genética , Miocárdio/patologia , Receptores do Ácido Retinoico/genética , Animais , Ecocardiografia , Feminino , Dosagem de Genes , Expressão Gênica , Marcação de Genes/métodos , Coração/embriologia , Coração/crescimento & desenvolvimento , Masculino , Camundongos , Camundongos Transgênicos , Cadeias Pesadas de Miosina/genética , Regiões Promotoras Genéticas , Função Ventricular Esquerda
15.
Proc Natl Acad Sci U S A ; 93(13): 6258-63, 1996 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-8692802

RESUMO

Tissue factor (TF) is the cellular receptor for coagulation factor VI/VIIa and is the membrane-bound glycoprotein that is generally viewed as the primary physiological initiator of blood coagulation. To define in greater detail the physiological role of TF in development and hemostasis, the TF gene was disrupted in mice. Mice heterozygous for the inactivated TF allele expressed approximately half the TF activity of wild-type mice but were phenotypically normal. However, homozygous TF-/- pups were never born in crosses between heterozygous mice. Analysis of mid-gestation embryos showed that TF-/- embryos die in utero between days 8.5 and 10.5. TF-/- embryos were morphologically distinct from their TF+/+ and TF+/- littermates after day 9.5 in that they were pale, edematous, and growth retarded. Histological studies showed that early organogenesis was normal. The initial failure in TF-/- embryos appeared to be hemorrhaging, leading to the leakage of embryonic red cells from both extraembryonic and embryonic vessels. These studies indicate that TF plays an indispensable role in establishing and/or maintaining vascular integrity in the developing embryo at a time when embryonic and extraembryonic vasculatures are fusing and blood circulation begins.


Assuntos
Transtornos da Coagulação Sanguínea/complicações , Morte Fetal/etiologia , Hemorragia , Tromboplastina/deficiência , Animais , Sequência de Bases , Primers do DNA , Genes Letais , Heterozigoto , Homozigoto , Camundongos , Dados de Sequência Molecular , Fenótipo , Tromboplastina/genética
16.
Circ Res ; 78(5): 790-8, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-8620598

RESUMO

During fetal development, a specialized vessel the ductus arteriosus, shunts blood from the pulmonary artery to the aorta, thus bypassing the lungs. The ductus differs primarily from the great vessels in that it is a muscular rather than an elastic artery, and the etiology of this differential development remains controversial. We present evidence that retinoic acid (RA) may contribute to the unique muscle phenotype of the ductus arteriosus. Using a transgenic mouse carrying an RA response element-lacZ transgene that expresses beta-galactosidase (beta-gal) in response to endogenous RA signals during embryonic and fetal development, we observe a strong beta-gal signal in the ductus arteriosus. By immunofluorescence, this signal colocalizes with the expression of the adult-specific smooth muscle myosin heavy chain isoform, SM2. The beta-gal signal is present throughout fetal development and persists in the neonate until the ductus arteriosus is completely closed. beta-Gal-positive cells are first detected by immunofluorescence at 13.5 days postcoitum (dpc) in the mesenchyme surrounding the ductus. By 15.5 dpc, very intense beta-gal staining localizes to the ductus arteriosus but is absent or minimal in the pulmonary trunk and aortic arch; by 17.5 dpc, the smooth muscle layers of the tunica media in the ductus arteriosus exhibit positive beta-gal staining. Immunostaining with antibodies against smooth muscle myosins shows that, while SM1 is expressed in all embryonic vessels, SM2 is precociously expressed in the ductus arteriosus. Furthermore, SM2 expression can be detected in the ductus as early as 15.5 dpc. In the neonate, the beta-gal signal persists in the smooth muscle layer of the ductus and immunostaining colocalizes with SM2 expression. These data suggest that RA may play a role in inducing and maintaining smooth muscle differentiation in the developing ductus arteriosus and may promote precocious expression of the adult vascular phenotype.


Assuntos
Canal Arterial/embriologia , Músculo Liso Vascular/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Transdução de Sinais , Tretinoína/metabolismo , Animais , Sequência de Bases , Desenvolvimento Embrionário e Fetal , Feminino , Humanos , Imuno-Histoquímica , Recém-Nascido , Isomerismo , Camundongos , Camundongos Transgênicos , Sondas Moleculares/genética , Dados de Sequência Molecular , Músculo Liso Vascular/embriologia , Músculo Liso Vascular/fisiologia , Fenótipo , Gravidez , Vitamina A/sangue , Deficiência de Vitamina A/complicações
17.
Circ Res ; 78(3): 504-9, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8593710

RESUMO

The structure-function relationships of the sarcomeric proteins in the mammalian cardiac compartment remain ill-defined because of the lack of a suitable model in which they can be readily manipulated or exchanged in vivo. To establish the validity of the transgenic paradigm for remodeling the mammalian heart, the murine alpha -cardiac myosin heavy chain gene promoter was used to express a ventricular myosin light chain-2 transgene (MLC2v) in both the atria and ventricles of the adult animal. Expression resulted in high levels of the transgene's transcript in both compartments. In the ventricle, the transgene was expressed against the background expression of the normal isoform. In the atrium, the transgene's expression would be ectopic, in that normally, MLC2v expression is restricted to the ventricle. Ectopic expression of the transgene in the atria resulted in a complete replacement of the atrial myosin light chain-2 protein isoform, although the endogenous isoform's steady state transcript levels were unchanged. In contrast, ventricular expression of the transgene had no effect at the protein level, despite an eightfold increase in MLC2v transcript levels. The data show that sarcomeric protein stoichiometry is maintained rigorously via posttransciptional regulation and that protein replacement can be achieved through a single transgenic manipulation.


Assuntos
Coração/fisiologia , Contração Miocárdica/genética , Miocárdio/metabolismo , Animais , Sequência de Bases , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Transgenes
19.
Dev Dyn ; 204(1): 1-12, 1995 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8563020

RESUMO

Retinoid-dependent gene expression accompanies the emergence of distinct regions and cell classes in the mouse spinal cord around midgestation. We asked whether changes in the expression of retinoid signaling molecules and retinoid-responsive genes reflect the establishment of this regional and cellular diversity. At E10.5, retinoic acid (RA) receptors (RAR)alpha, RAR beta, the retinoid X receptor (RXR) gamma, cellular RA binding protein (CRABP)I, CRAPBII, and cellular retinol binding protein (CRBP)I mRNAs are found throughout the entire anterior-posterior (AP) axis of the cord, as is RA (Colbert et al. [1993] Proc. Natl. Acad. Sci. U.S.A. 90:6572-6576) and RA-sensitive transgene expression (Balkan et al. [1992] Proc. Natl. Acad. Sci. U.S.A. 89:3347-3351). At E12.5, RA, transgene expression, and RAR beta become restricted to the cervical and lumbar cord. RAR alpha, CRABPI, and RXR gamma, however, are found throughout the AP extent. CRABPII and CRBPI, although expanded within the cervical and lumbar regions, are also found throughout the AP axis. Thus, several retinoid signaling molecules continue to be expressed beyond distinct regions of the spinal cord where RA is available and some RA-responsive genes are either restricted or enhanced. Exogenous RA can activate a more widespread response resulting in ectopic transgene and RAR beta expression in the thoracic and sacral cord. Not all RA-sensitive genes, however, respond; CRABPII and CRBPI expression patterns are unchanged. Finally, not every cell within the normal or exogenously induced domains of RA-dependent gene expression responds to RA, nor does every cell express RA receptors or binding proteins. Thus, regional and cellular differences in the distribution of the known retinoid receptors and binding proteins do not predict absolutely where or whether retinoid sensitive genes will be expressed or where retinoids will be available in the developing spinal cord. Instead, retinoid-mediated gene expression in the cervical and lumbar cord seems to reflect retinoid responses that rely both on the local availability of retinoids, the identity of the responding gene, and an indeterminate array of retinoid signaling molecules.


Assuntos
Camundongos Transgênicos/embriologia , Transdução de Sinais/genética , Medula Espinal/embriologia , Tretinoína/metabolismo , Animais , Indução Embrionária/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hibridização In Situ , Camundongos , Ligação Proteica/fisiologia , RNA Mensageiro/análise , Receptores do Ácido Retinoico/metabolismo , Receptores do Ácido Retinoico/fisiologia , Medula Espinal/citologia , Medula Espinal/ultraestrutura
20.
Development ; 121(3): 671-9, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7720575

RESUMO

The cytoplasmic retinoic acid (RA)-binding protein CRABP-II is expressed widely throughout early morphogenesis in mouse embryo, but its expression becomes more restricted as organogenesis progresses. CRABP-II expression remains strong in the developing limb bud suggesting a role for this protein in limb patterning. Here, we show that the CRABP-II promoter can direct expression of a lacZ transgene in a specific posterior domain during limb bud development. In order to investigate in more detail the role played by CRABP-II in RA signal transduction, we have also generated mice homozygous for a null mutation of this gene. CRABPII-/- mice are viable and fertile but show a developmental defect of the forelimb, specifically an additional, postaxial digit. This digit is generally, but not exclusively, limited to a single forepaw of an individual animal. The penetrance of the phenotype varies according to the genetic background, occurring most frequently on the inbred 129Sv background (50%), less frequently on the C57Bl/6 background (30%) and rarely on the outbred CD1 background (10%). This developmental abnormality implies a role for CRABP-II in normal patterning of the limb.


Assuntos
Membro Anterior/anormalidades , Polidactilia/genética , Receptores do Ácido Retinoico/genética , Animais , Expressão Gênica , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Morfogênese/efeitos dos fármacos , Morfogênese/genética , Fenótipo , Polidactilia/embriologia , Regiões Promotoras Genéticas , Transdução de Sinais , Tretinoína/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...