Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-32914013

RESUMO

PURPOSE: Next-generation sequencing (NGS) oncology panels are becoming integral in hospital and academic settings to guide patient treatment and enrollment in clinical trials. Although NGS technologies have revolutionized decision-making for cancer therapeutics, physicians may face many challenges in parsing and prioritizing NGS-based test results to determine the best course of treatment for individual patients. On January 29, 2018, the US Food and Drug Administration held a public workshop entitled, "Weighing the Evidence: Variant Classification and Interpretation in Precision Oncology." Here, we discuss the presentations and discussion highlights across the four sessions of the workshop. METHODS: The goal of the public workshop was to engage stakeholders and solicit input from experts in precision oncology to discuss the integration of complex NGS data into patient management and regulatory innovation within the precision oncology community. The US Food and Drug Administration gathered representatives from academia, industry, patient advocacy, government, and professional organizations for a series of presentations followed by panel discussions. After the workshop, the transcript and speaker presentation slides were reviewed and summarized for manuscript preparation. RESULTS: Speakers and panelists provided diverse perspectives on the integration of NGS technology into patient care for oncology and on the complexities that surround data interpretation and sharing. Discussions highlighted the challenges with standardization for variant classification while expressing the utility of consensus recommendations among stakeholders in oncology for driving innovation in the era of precision medicine. CONCLUSION: As precision medicine advances, clear communication within the field of precision oncology will be key to creating an environment that facilitates the generation and sharing of data that have value to patients.

2.
Clin Transl Sci ; 11(3): 267-276, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29498218

RESUMO

The high-content interrogation of single cells with platforms optimized for the multiparameter characterization of cells in liquid and solid biopsy samples can enable characterization of heterogeneous populations of cells ex vivo. Doing so will advance the diagnosis, prognosis, and treatment of cancer and other diseases. However, it is important to understand the unique issues in resolving heterogeneity and variability at the single cell level before navigating the validation and regulatory requirements in order for these technologies to impact patient care. Since 2013, leading experts representing industry, academia, and government have been brought together as part of the Foundation for the National Institutes of Health (FNIH) Biomarkers Consortium to foster the potential of high-content data integration for clinical translation.


Assuntos
Implementação de Plano de Saúde/métodos , Neoplasias/diagnóstico , Análise de Célula Única/métodos , Pesquisa Translacional Biomédica/métodos , Biópsia/métodos , Biópsia/normas , Implementação de Plano de Saúde/organização & administração , Humanos , National Institutes of Health (U.S.)/organização & administração , Neoplasias/patologia , Prognóstico , Análise de Célula Única/normas , Estados Unidos , Estudos de Validação como Assunto
3.
Clin Pharmacol Ther ; 104(2): 282-289, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29473145

RESUMO

Advances in our understanding of the molecular underpinnings of disease have spurred the development of targeted therapies and the use of precision medicine approaches in patient care. While targeted therapies have improved our capability to provide effective treatments to patients, they also present additional challenges to drug development and benefit-risk assessment such as identifying the subset(s) of patients likely to respond to the drug, assessing heterogeneity in response across molecular subsets of a disease, and developing diagnostic tests to identify patients for treatment. These challenges are particularly difficult to address when targeted therapies are developed to treat diseases with multiple molecular subtypes that occur at low frequencies. To help address these challenges, the US Food and Drug Administration recently published a draft guidance entitled "Developing Targeted Therapies in Low-Frequency Molecular Subsets of a Disease." Here we provide additional information on specific aspects of targeted therapy development in diseases with low-frequency molecular subsets.


Assuntos
Frequência do Gene , Predisposição Genética para Doença , Terapia de Alvo Molecular/métodos , Taxa de Mutação , Medicina de Precisão/métodos , Animais , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Medicina Baseada em Evidências , Humanos , Fenótipo , Estados Unidos , United States Food and Drug Administration
5.
Cerebellum ; 11(1): 41-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22548229

RESUMO

Granule neurons have a central role in cerebellar function via their synaptic interactions with other neuronal cell types both within and outside this structure. Establishment of these synaptic connections and its control is therefore essential to their function. Both intrinsic as well as environmental mechanisms are required for neuronal development and formation of neuronal circuits, and a key but poorly understood question is how these various events are coordinated and integrated in maturing neurons. In this review, we summarize recent work on the role of the Nuclear Factor I family in the transcriptional programming of cerebellar granule neuron maturation and synapse formation. In particular, we describe (1) the involvement of this family of factors in key developmental steps occurring throughout postmitotic granule neuron development, including dendrite and synapse formation and synaptic receptor expression, and (2) the mediation of these actions by critical downstream gene targets that control cell-cell interactions. These findings illustrate how Nuclear Factor I proteins and their regulons function as a "bridge" between cell-intrinsic and cell-extrinsic interactions to control multiple phases of granule neuron development.


Assuntos
Comunicação Celular/fisiologia , Diferenciação Celular/fisiologia , Córtex Cerebelar/crescimento & desenvolvimento , Córtex Cerebelar/metabolismo , Grânulos Citoplasmáticos/metabolismo , Fatores de Transcrição NFI/fisiologia , Neurogênese/fisiologia , Neurônios/metabolismo , Animais , Córtex Cerebelar/citologia , Grânulos Citoplasmáticos/fisiologia , Humanos , Fatores de Transcrição NFI/metabolismo , Vias Neurais/citologia , Vias Neurais/crescimento & desenvolvimento , Vias Neurais/metabolismo , Neurônios/citologia , Neurônios/fisiologia
6.
Dev Biol ; 354(2): 242-52, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21513708

RESUMO

The Nuclear factor I (NFI) transcription factor family consists of four genes (Nfia, Nfib, Nfic and Nfix) that regulate the development of multiple organ systems in mice and humans. Nfib is expressed in both lung mesenchyme and epithelium and mice lacking Nfib have severe lung maturation defects and die at birth. Here we continue our analysis of the phenotype of Nfib⁻/⁻ lungs and show that Nfib specifically in lung mesenchyme controls late epithelial and mesenchymal cell proliferation and differentiation. There are more PCNA, BrdU, PHH3 and Ki67 positive cells in Nfib⁻/⁻ lungs than in wild type lungs at E18.5 and this increase in proliferation marker expression is seen in both epithelial and mesenchymal cells. The loss of Nfib in all lung cells decreases the expression of markers for alveolar epithelial cells (Aqp5 and Sftpc), Clara cells (Scgb1a1) and ciliated cells (Foxj1) in E18.5 lungs. To test for a specific role of Nfib in lung mesenchyme we generated and analyzed Nfib(flox/flox), Dermo1-Cre mice. Loss of Nfib only in mesenchyme results in decreased Aqp5, Sftpc and Foxj1 expression, increased cell proliferation, and a defect in sacculation similar to that seen in Nfib⁻/⁻ mice. In contrast, mesenchyme specific loss of Nfib had no effect on the expression of Scgb1a1 in the airway. Microarray and QPCR analyses indicate that the loss of Nfib in lung mesenchyme affects the expression of genes associated with extracellular matrix, cell adhesion and FGF signaling which could affect distal lung maturation. Our data indicate that mesenchymal Nfib regulates both mesenchymal and epithelial cell proliferation through multiple pathways and that mesenchymal NFI-B-mediated signals are essential for the maturation of distal lung epithelium.


Assuntos
Diferenciação Celular , Proliferação de Células , Pulmão/crescimento & desenvolvimento , Fatores de Transcrição NFI/metabolismo , Mucosa Respiratória/citologia , Animais , Aquaporina 5/metabolismo , Bromodesoxiuridina/metabolismo , Epitélio/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intercelular , Antígeno Ki-67/metabolismo , Pulmão/citologia , Pulmão/embriologia , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Peptídeos/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteína C Associada a Surfactante Pulmonar , Transdução de Sinais , Uteroglobina/metabolismo
7.
J Neurosci Res ; 88(2): 258-65, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19658195

RESUMO

Recent studies have shown that the nuclear factor I (NFI) family controls multiple stages of the postmitotic differentiation of cerebellar granule neurons (CGNs). Regulation of cell-cell signaling is an integral part of this NFI program, which involves expression of the cell adhesion molecules N cadherin and ephrin B1 throughout postmitotic CGN development. Here, we identify two additional downstream targets of NFI that are involved in extracellular CGN interactions. The cell adhesion molecule Tag-1 is highly enriched in CGNs undergoing parallel fiber formation and is down-regulated prior to onset of radial migration. We found that Tag-1 expression was strongly reduced by NFI dominant repression in immature primary CGNs and in the cerebella of E18 Nfib-null mice. Transient transfection and chromatin immunoprecipitation suggested that the Tag-1 gene is directly regulated by NFI. Furthermore, functional, Nfi knockout and chromatin immunoprecipitation studies implicated Wnt7a as a direct target of NFI in maturing CGNs. Wnt7a is secreted by developing CGNs and is required for maturation of mossy fiber-CGN synaptic rosettes. Consistent with this, synapsin I was greatly reduced within the internal granule cell layer of P17 Nfia-null mice. These findings indicated that NFI controls CGN postmitotic maturation through a combination of extracellular signaling molecules that operate either continuously to regulate multiple stages of development (N cadherin and ephrin B1) or primarily at early (Tag-1) or late (Wnt7a) maturation steps. They also illustrate the importance of NFI as a critical link between cell-intrinsic mechanisms and cell-cell interactions in the development of the mouse cerebellum.


Assuntos
Cerebelo/fisiologia , Fatores de Transcrição NFI/genética , Neurônios/fisiologia , Regulon , Animais , Caderinas/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Comunicação Celular/fisiologia , Células Cultivadas , Cerebelo/embriologia , Cerebelo/crescimento & desenvolvimento , Cromatina/metabolismo , Contactina 2 , Efrina-B1/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitose , Fatores de Transcrição NFI/metabolismo , Transdução de Sinais , Fatores de Tempo , Proteínas Wnt/metabolismo
8.
J Comp Neurol ; 513(1): 98-112, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19107796

RESUMO

Transcription factors of the Nuclear Factor I (Nfi) family are important for the development of specific neuronal and glial populations in the nervous system. One such population, the neurons of the basilar pontine nuclei, expresses high levels of Nfi proteins, and the pontine nuclei are greatly reduced in mice lacking a functional Nfib gene. Pontine neurons, along with other precerebellar neurons that populate the hindbrain, arise from precursors in the lower rhombic lip and migrate anteroventrally to reach their final location. Using immunohistochemistry, we find that NFI-B expression is specific for mossy fiber populations of the precerebellar system. Analysis of the Nfib(-/-) hindbrain indicates that the development of the basilar pontine nuclei is delayed, with pontine neurons migrating 1-2 days later than in control animals, and that significantly fewer pontine neurons are produced. While the mossy fiber nuclei of the caudal medulla do form, they also exhibit a developmental delay. Nfia and Nfix null mice exhibit no apparent pontine phenotype, implying specificity in the action of NFI family members. Collectively, these data demonstrate that Nfib plays an important role in the generation of precerebellar mossy fiber neurons, and may do so at least in part by regulating neurogenesis.


Assuntos
Córtex Cerebelar , Fatores de Transcrição NFI/metabolismo , Vias Neurais , Ponte , Animais , Córtex Cerebelar/anormalidades , Córtex Cerebelar/anatomia & histologia , Córtex Cerebelar/embriologia , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Transcrição NFI/genética , Vias Neurais/anormalidades , Vias Neurais/anatomia & histologia , Vias Neurais/embriologia , Neurônios/citologia , Neurônios/fisiologia , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , Fenótipo , Ponte/anormalidades , Ponte/anatomia & histologia , Ponte/embriologia , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Rombencéfalo/anormalidades , Rombencéfalo/anatomia & histologia , Rombencéfalo/embriologia , Rombencéfalo/metabolismo
9.
BMC Dev Biol ; 8: 52, 2008 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-18477394

RESUMO

BACKGROUND: The Nuclear Factor I (NFI) multi-gene family encodes site-specific transcription factors essential for the development of a number of organ systems. We showed previously that Nfia-deficient mice exhibit agenesis of the corpus callosum and other forebrain defects; Nfib-deficient mice have defects in lung maturation and show callosal agenesis and forebrain defects resembling those seen in Nfia-deficient animals, while Nfic-deficient mice have defects in tooth root formation. Recently the Nfix gene has been disrupted and these studies indicated that there were largely uncharacterized defects in brain and skeletal development in Nfix-deficient mice. RESULTS: Here we show that disruption of Nfix by Cre-recombinase mediated excision of the 2nd exon results in defects in brain development that differ from those seen in Nfia and Nfib KO mice. In particular, complete callosal agenesis is not seen in Nfix-/- mice but rather there appears to be an overabundance of aberrant Pax6- and doublecortin-positive cells in the lateral ventricles of Nfix-/- mice, increased brain weight, expansion of the cingulate cortex and entire brain along the dorsal ventral axis, and aberrant formation of the hippocampus. On standard lab chow Nfix-/- animals show a decreased growth rate from ~P8 to P14, lose weight from ~P14 to P22 and die at ~P22. If their food is supplemented with a soft dough chow from P10, Nfix-/- animals show a lag in weight gain from P8 to P20 but then increase their growth rate. A fraction of the animals survive to adulthood and are fertile. The weight loss correlates with delayed eye and ear canal opening and suggests a delay in the development of several epithelial structures in Nfix-/- animals. CONCLUSION: These data show that Nfix is essential for normal brain development and may be required for neural stem cell homeostasis. The delays seen in eye and ear opening and the brain morphology defects appear independent of the nutritional deprivation, as rescue of perinatal lethality with soft dough does not eliminate these defects.


Assuntos
Encéfalo/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Genes Controladores do Desenvolvimento , Fatores de Transcrição NFI/genética , Animais , Western Blotting , Córtex Cerebral/crescimento & desenvolvimento , Ventrículos Cerebrais/crescimento & desenvolvimento , Embrião de Mamíferos , Células-Tronco Embrionárias , Feminino , Marcação de Genes , Hipocampo/crescimento & desenvolvimento , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Reação em Cadeia da Polimerase
10.
J Neurosci ; 27(23): 6115-27, 2007 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-17553984

RESUMO

A central question is how various stages of neuronal development are integrated as a differentiation program. Here we show that the nuclear factor I (NFI) family of transcriptional regulators is expressed and functions throughout the postmitotic development of cerebellar granule neurons (CGNs). Expression of an NFI dominant repressor in CGN cultures blocked axon outgrowth and dendrite formation and decreased CGN migration. Inhibition of NFI transactivation also disrupted extension and fasciculation of parallel fibers as well as CGN migration to the internal granule cell layer in cerebellar slices. In postnatal day 17 Nfia-deficient mice, parallel fibers were greatly diminished and disoriented, CGN dendrite formation was dramatically impaired, and migration from the external germinal layer (EGL) was retarded. Axonal marker expression also was disrupted within the EGL of embryonic day 18 Nfib-null mice. NFI regulation of axon extension was observed under conditions of homotypic cell contact, implicating cell surface proteins as downstream mediators of its actions in CGNs. Consistent with this, the cell adhesion molecules ephrin B1 and N-cadherin were identified as NFI gene targets in CGNs using inhibitor and Nfi mutant analysis as well as chromatin immunoprecipitation. Functional inhibition of ephrin B1 or N-cadherin interfered with CGN axon extension and guidance, migration, and dendritogenesis in cell culture as well as in situ. These studies define NFI as a key regulator of postmitotic CGN development, in particular of axon formation, dendritogenesis, and migratory behavior. Furthermore, they reveal how a single transcription factor family can control and integrate multiple aspects of neuronal differentiation through the regulation of cell adhesion molecules.


Assuntos
Moléculas de Adesão Celular/fisiologia , Cerebelo/citologia , Cerebelo/crescimento & desenvolvimento , Fatores de Transcrição NFI/fisiologia , Animais , Movimento Celular/fisiologia , Células Cultivadas , Cerebelo/fisiologia , Drosophila , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ratos
11.
Cereb Cortex ; 17(7): 1531-41, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16959869

RESUMO

The laminar and area patterning of the mammalian neocortex are two organizing principles that define its functional architecture. Members of the immunoglobulin (Ig) superfamily of cell adhesion molecules influence neural development by regulating cell adhesion, migration, and process growth. Here we describe the dynamic expression of the unique Ig-containing cell adhesion molecule, MAM domain-containing glycosylphosphatidylinositol anchor 1 (MDGA1), during forebrain development in mice and compare it with other markers. We show that MDGA1 is a layer-specific marker and an area-specific marker, being expressed in layers 2/3 throughout the neocortex, but within the primary somatosensory area (S1), MDGA1 is also uniquely expressed in layers 4 and 6a. Comparisons with other markers, including cadherins, serotonin, cytochrome oxidase, ROR beta, and COUP-TF1, reveal unique features of patterned expression of MDGA1 within cortex and S1 barrels. Further, our findings indicate that at earlier stages of development, MDGA1 is expressed by Reelin- and Tbr1-positive Cajal-Retzius neurons that originate from multiple sources outside of neocortex and emigrate into it. At even earlier stages, MDGA1 is expressed by the earliest diencephalic and mesencephalic neurons, which appear to migrate from a MDGA1-positive domain of progenitors in the diencephalon and form a "preplate." These findings show that MDGA1 is a unique marker for studies of cortical lamination and area patterning and together with recent reports suggest that MDGA1 has critical functions in forebrain/midbrain development.


Assuntos
Moléculas de Adesão Celular/metabolismo , Rede Nervosa/citologia , Rede Nervosa/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Prosencéfalo/citologia , Prosencéfalo/metabolismo , Animais , Biomarcadores/metabolismo , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Proteínas Ligadas por GPI , Camundongos , Camundongos Endogâmicos C57BL , Moléculas de Adesão de Célula Nervosa , Neurônios/classificação , Proteína Reelina , Distribuição Tecidual
12.
Brain Res ; 1117(1): 12-7, 2006 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-16956587

RESUMO

Several forms of congenital muscular dystrophy result from mutations in glycosyltransferases that modify alpha-dystroglycan. As pontine hypoplasia has been reported in some clinical cases of congenital muscular dystrophy, we have begun to examine whether these glycosyltransferases are required for the normal development of the basilar pons, one of several precerebellar nuclei of the hindbrain. In veils (Large(vls)) mice, which carry a loss-of-function mutation in the Large glycosyltransferase gene, the basilar pons is absent. Instead, ectopic clusters of pontine neurons are found lateral to their normal site, suggesting that these neurons are unable to migrate to their appropriate site. Two other precerebellar nuclei, the lateral reticular nucleus and the inferior olive, are present in Large(vls) mice. In addition, the basilar pons forms normally in dystrophin-deficient mice. These results demonstrate that the Large glycosyltransferase but not dystrophin is required for normal basilar pontine development.


Assuntos
Movimento Celular/genética , Glicosiltransferases/genética , Malformações do Sistema Nervoso/enzimologia , Neurônios/enzimologia , Ponte/anormalidades , Ponte/enzimologia , Animais , Diferenciação Celular/genética , Cerebelo/anormalidades , Cerebelo/enzimologia , Cerebelo/fisiopatologia , Distroglicanas/metabolismo , Distrofina/deficiência , Distrofina/genética , Camundongos , Camundongos Mutantes Neurológicos , Mutação/genética , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/fisiopatologia , Vias Neurais/anormalidades , Vias Neurais/enzimologia , Vias Neurais/fisiopatologia , Ponte/fisiopatologia
13.
Mol Cell Biol ; 25(2): 685-98, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15632069

RESUMO

The phylogenetically conserved nuclear factor I (NFI) gene family encodes site-specific transcription factors essential for the development of a number of organ systems. We showed previously that Nfia-deficient mice exhibit agenesis of the corpus callosum and other forebrain defects, whereas Nfic-deficient mice have agenesis of molar tooth roots and severe incisor defects. Here we show that Nfib-deficient mice possess unique defects in lung maturation and exhibit callosal agenesis and forebrain defects that are similar to, but more severe than, those seen in Nfia-deficient animals. In addition, loss of Nfib results in defects in basilar pons formation and hippocampus development that are not seen in Nfia-deficient mice. Heterozygous Nfib-deficient animals also exhibit callosal agenesis and delayed lung maturation, indicating haploinsufficiency at the Nfib locus. The similarity in brain defects in Nfia- and Nfib-deficient animals suggests that these two genes may cooperate in late fetal forebrain development, while Nfib is essential for late fetal lung maturation and development of the pons.


Assuntos
Encéfalo/crescimento & desenvolvimento , Pulmão/crescimento & desenvolvimento , Proteínas/metabolismo , Agenesia do Corpo Caloso , Animais , Biomarcadores , Encéfalo/anormalidades , Diferenciação Celular , Embrião de Mamíferos/anatomia & histologia , Embrião de Mamíferos/fisiologia , Feminino , Marcação de Genes , Idade Gestacional , Humanos , Pulmão/anormalidades , Pulmão/anatomia & histologia , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição NFI , Gravidez , Proteínas/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
14.
Mol Cell Neurosci ; 25(2): 263-74, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15019943

RESUMO

We recently used a differential display PCR screen to identify secreted and transmembrane proteins that are highly expressed in the developing rat basilar pons, a prominent ventral hindbrain nucleus used as a model for studies of neuronal migration, axon outgrowth, and axon-target recognition. Here we describe cloning and characterization of one of these molecules, now called MDGA1, and a closely related homologue, MDGA2. Analyses of the full-length coding region of MDGA1 and MDGA2 indicate that they encode proteins that comprise a novel subgroup of the Ig superfamily and have a unique structural organization consisting of six immunoglobulin (Ig)-like domains followed by a single MAM domain. Biochemical characterization demonstrates that MDGA1 and MDGA2 proteins are highly glycosylated, and that MDGA1 is tethered to the cell membrane by a GPI anchor. The MDGAs are differentially expressed by subpopulations of neurons in both the central and peripheral nervous systems, including neurons of the basilar pons, inferior olive, cerebellum, cerebral cortex, olfactory bulb, spinal cord, and dorsal root and trigeminal ganglia. Little or no MDGA expression is detected outside of the nervous system of developing rats. The similarity of MDGAs to other Ig-containing molecules and their temporal-spatial patterns of expression within restricted neuronal populations, for example migrating pontine neurons and D1 spinal interneurons, suggest a role for these novel proteins in regulating neuronal migration, as well as other aspects of neural development, including axon guidance.


Assuntos
Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Cones de Crescimento/metabolismo , Imunoglobulinas/genética , Glicoproteínas de Membrana/genética , Sistema Nervoso/embriologia , Sequência de Aminoácidos/genética , Animais , Sequência de Bases/genética , Sistema Nervoso Central/citologia , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/metabolismo , DNA Complementar/análise , DNA Complementar/genética , Feto , Proteínas Ligadas por GPI , Regulação da Expressão Gênica no Desenvolvimento/genética , Glicosilação , Cones de Crescimento/ultraestrutura , Imunoglobulinas/biossíntese , Imunoglobulinas/isolamento & purificação , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/isolamento & purificação , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Proteínas de Membrana/isolamento & purificação , Dados de Sequência Molecular , Fatores de Crescimento Neural/biossíntese , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/isolamento & purificação , Sistema Nervoso/citologia , Sistema Nervoso/metabolismo , Moléculas de Adesão de Célula Nervosa , Sistema Nervoso Periférico/citologia , Sistema Nervoso Periférico/embriologia , Sistema Nervoso Periférico/metabolismo , Ponte/citologia , Ponte/embriologia , Ponte/metabolismo , Estrutura Terciária de Proteína/fisiologia , Ratos , Homologia de Sequência de Aminoácidos
15.
J Cell Biol ; 156(5): 879-92, 2002 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-11864995

RESUMO

The nonreceptor tyrosine kinase encoded by the c-Abl gene has the unique feature of an F-actin binding domain (FABD). Purified c-Abl tyrosine kinase is inhibited by F-actin, and this inhibition can be relieved through mutation of its FABD. The c-Abl kinase is activated by physiological signals that also regulate the actin cytoskeleton. We show here that c-Abl stimulated the formation of actin microspikes in fibroblasts spreading on fibronectin. This function of c-Abl is dependent on kinase activity and is not shared by c-Src tyrosine kinase. The Abl-dependent F-actin microspikes occurred under conditions where the Rho-family GTPases were inhibited. The FABD-mutated c-Abl, which is active in detached fibroblasts, stimulated F-actin microspikes independent of cell attachment. Moreover, FABD-mutated c-Abl stimulated the formation of F-actin branches in neurites of rat embryonic cortical neurons. The reciprocal regulation between F-actin and the c-Abl tyrosine kinase may provide a self-limiting mechanism in the control of actin cytoskeleton dynamics.


Assuntos
Actinas/metabolismo , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Córtex Cerebral/embriologia , Citoesqueleto/enzimologia , Neuritos/metabolismo , Proteínas Proto-Oncogênicas c-abl/deficiência , Animais , Benzamidas , Adesão Celular/fisiologia , Tamanho Celular/fisiologia , Córtex Cerebral/citologia , Córtex Cerebral/enzimologia , Citoesqueleto/ultraestrutura , Matriz Extracelular/metabolismo , Fibronectinas/farmacologia , Imunofluorescência , Mesilato de Imatinib , Camundongos , Neuritos/enzimologia , Neuritos/ultraestrutura , Piperazinas , Proteínas Proto-Oncogênicas c-abl/genética , Pirimidinas/farmacologia , Ratos , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...