Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Nat Med ; 30(5): 1406-1415, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38745011

RESUMEN

GRN mutations cause progranulin haploinsufficiency, which eventually leads to frontotemporal dementia (FTD-GRN). PR006 is an investigational gene therapy delivering the granulin gene (GRN) using an adeno-associated virus serotype 9 (AAV9) vector. In non-clinical studies, PR006 transduced neurons derived from induced pluripotent stem cells of patients with FTD-GRN, resulted in progranulin expression and improvement of lipofuscin, lysosomal and neuroinflammation pathologies in Grn-knockout mice, and was well tolerated except for minimal, asymptomatic dorsal root ganglionopathy in non-human primates. We initiated a first-in-human phase 1/2 open-label trial. Here we report results of a pre-specified interim analysis triggered with the last treated patient of the low-dose cohort (n = 6) reaching the 12-month follow-up timepoint. We also include preliminary data from the mid-dose cohort (n = 7). Primary endpoints were safety, immunogenicity and change in progranulin levels in cerebrospinal fluid (CSF) and blood. Secondary endpoints were Clinical Dementia Rating (CDR) plus National Alzheimer's Disease Coordinating Center (NACC) Frontotemporal Lobar Degeneration (FTLD) rating scale and levels of neurofilament light chain (NfL). One-time administration of PR006 into the cisterna magna was generally safe and well tolerated. All patients developed treatment-emergent anti-AAV9 antibodies in the CSF, but none developed anti-progranulin antibodies. CSF pleocytosis was the most common PR006-related adverse event. Twelve serious adverse events occurred, mostly unrelated to PR006. Deep vein thrombosis developed in three patients. There was one death (unrelated) occurring 18 months after treatment. CSF progranulin increased after PR006 treatment in all patients; blood progranulin increased in most patients but only transiently. NfL levels transiently increased after PR006 treatment, likely reflecting dorsal root ganglia toxicity. Progression rates, based on the CDR scale, were within the broad ranges reported for patients with FTD. These data provide preliminary insights into the safety and bioactivity of PR006. Longer follow-up and additional studies are needed to confirm the safety and potential efficacy of PR006. ClinicalTrials.gov identifier: NCT04408625 .


Asunto(s)
Dependovirus , Demencia Frontotemporal , Terapia Genética , Progranulinas , Humanos , Demencia Frontotemporal/genética , Demencia Frontotemporal/terapia , Demencia Frontotemporal/líquido cefalorraquídeo , Progranulinas/genética , Terapia Genética/efectos adversos , Terapia Genética/métodos , Dependovirus/genética , Persona de Mediana Edad , Femenino , Masculino , Anciano , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/líquido cefalorraquídeo , Vectores Genéticos , Animales , Resultado del Tratamiento , Investigación Biomédica Traslacional , Ratones , Proteínas de Neurofilamentos/genética , Proteínas de Neurofilamentos/líquido cefalorraquídeo , Proteínas de Neurofilamentos/sangre
2.
Artículo en Inglés | MEDLINE | ID: mdl-32914013

RESUMEN

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.

3.
Clin Transl Sci ; 11(3): 267-276, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29498218

RESUMEN

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.


Asunto(s)
Implementación de Plan de Salud/métodos , Neoplasias/diagnóstico , Análisis de la Célula Individual/métodos , Investigación Biomédica Traslacional/métodos , Biopsia/métodos , Biopsia/normas , Implementación de Plan de Salud/organización & administración , Humanos , National Institutes of Health (U.S.)/organización & administración , Neoplasias/patología , Pronóstico , Análisis de la Célula Individual/normas , Estados Unidos , Estudios de Validación como Asunto
4.
Clin Pharmacol Ther ; 104(2): 282-289, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29473145

RESUMEN

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.


Asunto(s)
Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Terapia Molecular Dirigida/métodos , Tasa de Mutación , Medicina de Precisión/métodos , Animales , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Medicina Basada en la Evidencia , Humanos , Fenotipo , Estados Unidos , United States Food and Drug Administration
5.
Clin Pharmacol Ther ; 99(2): 198-207, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26536838

RESUMEN

High throughput molecular and functional profiling of patients is a key driver of precision medicine. DNA and RNA characterization has been enabled at unprecedented cost and scale through rapid, disruptive progress in sequencing technology, but challenges persist in data management and interpretation. We analyze the state-of-the-art of large-scale unbiased sequencing in drug discovery and development, including technology, application, ethical, regulatory, policy and commercial considerations, and discuss issues of LUS implementation in clinical and regulatory practice.


Asunto(s)
Bases de Datos Factuales/tendencias , Descubrimiento de Drogas/tendencias , Farmacogenética/tendencias , Bases de Datos Factuales/legislación & jurisprudencia , Bases de Datos Factuales/normas , Atención a la Salud/tendencias , Descubrimiento de Drogas/legislación & jurisprudencia , Descubrimiento de Drogas/normas , Pruebas Genéticas , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Farmacogenética/legislación & jurisprudencia , Farmacogenética/normas , Medicina de Precisión , Estados Unidos , United States Food and Drug Administration
8.
Cerebellum ; 11(1): 41-9, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22548229

RESUMEN

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.


Asunto(s)
Comunicación Celular/fisiología , Diferenciación Celular/fisiología , Corteza Cerebelosa/crecimiento & desarrollo , Corteza Cerebelosa/metabolismo , Gránulos Citoplasmáticos/metabolismo , Factores de Transcripción NFI/fisiología , Neurogénesis/fisiología , Neuronas/metabolismo , Animales , Corteza Cerebelosa/citología , Gránulos Citoplasmáticos/fisiología , Humanos , Factores de Transcripción NFI/metabolismo , Vías Nerviosas/citología , Vías Nerviosas/crecimiento & desarrollo , Vías Nerviosas/metabolismo , Neuronas/citología , Neuronas/fisiología
9.
Dev Biol ; 354(2): 242-52, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21513708

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Pulmón/crecimiento & desarrollo , Factores de Transcripción NFI/metabolismo , Mucosa Respiratoria/citología , Animales , Acuaporina 5/metabolismo , Bromodesoxiuridina/metabolismo , Epitelio/metabolismo , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular , Antígeno Ki-67/metabolismo , Pulmón/citología , Pulmón/embriología , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Péptidos/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteína C Asociada a Surfactante Pulmonar , Transducción de Señal , Uteroglobina/metabolismo
10.
J Neurosci Res ; 88(2): 258-65, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19658195

RESUMEN

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.


Asunto(s)
Cerebelo/fisiología , Factores de Transcripción NFI/genética , Neuronas/fisiología , Regulón , Animales , Cadherinas/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Comunicación Celular/fisiología , Células Cultivadas , Cerebelo/embriología , Cerebelo/crecimiento & desarrollo , Cromatina/metabolismo , Contactina 2 , Efrina-B1/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitosis , Factores de Transcripción NFI/metabolismo , Transducción de Señal , Factores de Tiempo , Proteínas Wnt/metabolismo
11.
J Comp Neurol ; 513(1): 98-112, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19107796

RESUMEN

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.


Asunto(s)
Corteza Cerebelosa , Factores de Transcripción NFI/metabolismo , Vías Nerviosas , Puente , Animales , Corteza Cerebelosa/anomalías , Corteza Cerebelosa/anatomía & histología , Corteza Cerebelosa/embriología , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción NFI/genética , Vías Nerviosas/anomalías , Vías Nerviosas/anatomía & histología , Vías Nerviosas/embriología , Neuronas/citología , Neuronas/fisiología , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Fenotipo , Puente/anomalías , Puente/anatomía & histología , Puente/embriología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Rombencéfalo/anomalías , Rombencéfalo/anatomía & histología , Rombencéfalo/embriología , Rombencéfalo/metabolismo
12.
BMC Dev Biol ; 8: 52, 2008 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-18477394

RESUMEN

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.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Genes del Desarrollo , Factores de Transcripción NFI/genética , Animales , Western Blotting , Corteza Cerebral/crecimiento & desarrollo , Ventrículos Cerebrales/crecimiento & desarrollo , Embrión de Mamíferos , Células Madre Embrionarias , Femenino , Marcación de Gen , Hipocampo/crecimiento & desarrollo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Reacción en Cadena de la Polimerasa
13.
J Neurosci ; 27(23): 6115-27, 2007 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-17553984

RESUMEN

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.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Cerebelo/citología , Cerebelo/crecimiento & desarrollo , Factores de Transcripción NFI/fisiología , Animales , Movimiento Celular/fisiología , Células Cultivadas , Cerebelo/fisiología , Drosophila , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ratas
14.
Cereb Cortex ; 17(7): 1531-41, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16959869

RESUMEN

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.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Red Nerviosa/citología , Red Nerviosa/metabolismo , Neuronas/citología , Neuronas/metabolismo , Prosencéfalo/citología , Prosencéfalo/metabolismo , Animales , Biomarcadores/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Proteínas Ligadas a GPI , Ratones , Ratones Endogámicos C57BL , Moléculas de Adhesión de Célula Nerviosa , Neuronas/clasificación , Proteína Reelina , Distribución Tisular
15.
Brain Res ; 1117(1): 12-7, 2006 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-16956587

RESUMEN

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.


Asunto(s)
Movimiento Celular/genética , Glicosiltransferasas/genética , Malformaciones del Sistema Nervioso/enzimología , Neuronas/enzimología , Puente/anomalías , Puente/enzimología , Animales , Diferenciación Celular/genética , Cerebelo/anomalías , Cerebelo/enzimología , Cerebelo/fisiopatología , Distroglicanos/metabolismo , Distrofina/deficiencia , Distrofina/genética , Ratones , Ratones Mutantes Neurológicos , Mutación/genética , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/fisiopatología , Vías Nerviosas/anomalías , Vías Nerviosas/enzimología , Vías Nerviosas/fisiopatología , Puente/fisiopatología
16.
Mol Cell Biol ; 25(2): 685-98, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15632069

RESUMEN

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.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Pulmón/crecimiento & desarrollo , Proteínas/metabolismo , Agenesia del Cuerpo Calloso , Animales , Biomarcadores , Encéfalo/anomalías , Diferenciación Celular , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Femenino , Marcación de Gen , Edad Gestacional , Humanos , Pulmón/anomalías , Pulmón/anatomía & histología , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción NFI , Embarazo , Proteínas/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
17.
Mol Cell Neurosci ; 25(2): 263-74, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15019943

RESUMEN

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.


Asunto(s)
Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Conos de Crecimiento/metabolismo , Inmunoglobulinas/genética , Glicoproteínas de Membrana/genética , Sistema Nervioso/embriología , Secuencia de Aminoácidos/genética , Animales , Secuencia de Bases/genética , Sistema Nervioso Central/citología , Sistema Nervioso Central/embriología , Sistema Nervioso Central/metabolismo , ADN Complementario/análisis , ADN Complementario/genética , Feto , Proteínas Ligadas a GPI , Regulación del Desarrollo de la Expresión Génica/genética , Glicosilación , Conos de Crecimiento/ultraestructura , Inmunoglobulinas/biosíntesis , Inmunoglobulinas/aislamiento & purificación , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/aislamiento & purificación , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Proteínas de la Membrana/aislamiento & purificación , Datos de Secuencia Molecular , Factores de Crecimiento Nervioso/biosíntesis , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/aislamiento & purificación , Sistema Nervioso/citología , Sistema Nervioso/metabolismo , Moléculas de Adhesión de Célula Nerviosa , Sistema Nervioso Periférico/citología , Sistema Nervioso Periférico/embriología , Sistema Nervioso Periférico/metabolismo , Puente/citología , Puente/embriología , Puente/metabolismo , Estructura Terciaria de Proteína/fisiología , Ratas , Homología de Secuencia de Aminoácido
18.
J Cell Biol ; 156(5): 879-92, 2002 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-11864995

RESUMEN

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.


Asunto(s)
Actinas/metabolismo , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Corteza Cerebral/embriología , Citoesqueleto/enzimología , Neuritas/metabolismo , Proteínas Proto-Oncogénicas c-abl/deficiencia , Animales , Benzamidas , Adhesión Celular/fisiología , Tamaño de la Célula/fisiología , Corteza Cerebral/citología , Corteza Cerebral/enzimología , Citoesqueleto/ultraestructura , Matriz Extracelular/metabolismo , Fibronectinas/farmacología , Técnica del Anticuerpo Fluorescente , Mesilato de Imatinib , Ratones , Neuritas/enzimología , Neuritas/ultraestructura , Piperazinas , Proteínas Proto-Oncogénicas c-abl/genética , Pirimidinas/farmacología , Ratas , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
19.
Mol Cell Neurosci ; 18(1): 1-12, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11461149

RESUMEN

The basilar pons, a major hindbrain nucleus involved in sensory-motor integration, has become a model system for studying long-distance neuronal migration, axon-target recognition by collateral branching, and the formation of patterned axonal projections. To identify genes potentially involved in these developmental events, we have performed a differential display PCR screen comparing RNA isolated from the developing basilar pons with RNA obtained from developing cerebellum and olfactory bulb, as well as the mature basilar pons. Using 400 different combinations of primers, we screened more than 11,000 labeled DNA fragments and identified 201 that exhibited higher expression in the basilar pons than in the control tissues. From these, 138 distinct gene fragments were cloned. The differential expression of a large subset of these fragments was confirmed using RNase protection assays. In situ hybridization analysis revealed that the expression of many of these genes is limited to the basilar pons and only a few other brain regions, suggesting that they may play specific roles in pontine development.


Asunto(s)
Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Puente/embriología , Puente/fisiología , Animales , Química Encefálica/genética , Clonación Molecular , Cartilla de ADN , Femenino , Embarazo , Ratas , Ratas Sprague-Dawley , Factores de Transcripción/genética
20.
J Biol Chem ; 273(35): 22825-32, 1998 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-9712917

RESUMEN

ARH-77 cells do not adhere to type I collagen and readily invade into collagen gels, but following expression of the transmembrane heparan sulfate proteoglycan syndecan-1, they bind collagen and fail to invade. We now show that cells transfected with syndecan-2 or syndecan-4 also bind collagen and are non-invasive. In contrast, cells transfected with the glycosylphosphatidylinositol-anchored proteoglycan glypican-1 do not bind to collagen and remain invasive, even though glypican- and syndecan-expressing cells have similar surface levels of heparan sulfate, and their proteoglycans have similar affinities for collagen. Analysis of cells expressing syndecan-1-glypican-1 chimeric proteoglycans reveals that inhibition of invasion requires the extracellular domain of syndecan but not its transmembrane or cytoplasmic domain. Surprisingly, cells bearing a chimera composed of the glypican extracellular domain fused to the syndecan transmembrane and cytoplasmic domains bind to collagen but remain invasive, implying that adhesion to collagen is not by itself sufficient to inhibit invasion. Apparently, the extracellular domain of syndecan-1, presumably by interacting with cell-surface signal transducing molecules, directly regulates complex cell behaviors such as motility and invasiveness. These results also show for the first time that syndecans and glypicans can have distinct functions, even when expressed by the same cell type.


Asunto(s)
Adhesión Celular/fisiología , Proteoglicanos de Heparán Sulfato/fisiología , Glicoproteínas de Membrana/fisiología , Invasividad Neoplásica/patología , Proteoglicanos/fisiología , Animales , Línea Celular , Colágeno , Proteoglicanos de Heparán Sulfato/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteoglicanos/metabolismo , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Relación Estructura-Actividad , Sindecano-1 , Sindecanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA