Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neurosci ; 41(8): 1650-1664, 2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33452226

RESUMEN

Promoting oligodendrocyte (OL) differentiation represents a promising option for remyelination therapy for treating the demyelinating disease multiple sclerosis (MS). The Wnt effector transcription factor 7-like 2 (TCF7l2) was upregulated in MS lesions and had been proposed to inhibit OL differentiation. Recent data suggest the opposite yet underlying mechanisms remain elusive. Here, we unravel a previously unappreciated function of TCF7l2 in controlling autocrine bone morphogenetic protein (BMP)4-mediated signaling. Disrupting TCF7l2 in mice of both sexes results in oligodendroglial-specific BMP4 upregulation and canonical BMP4 signaling activation in vivo Mechanistically, TCF7l2 binds to Bmp4 gene regulatory element and directly represses its transcriptional activity. Functionally, enforced TCF7l2 expression promotes OL differentiation by reducing autocrine BMP4 secretion and dampening BMP4 signaling. Importantly, compound genetic disruption demonstrates that oligodendroglial-specific BMP4 deletion rescues arrested OL differentiation elicited by TCF7l2 disruption in vivo Collectively, our study reveals a novel connection between TCF7l2 and BMP4 in oligodendroglial lineage and provides new insights into augmenting TCF7l2 for promoting remyelination in demyelinating disorders such as MS.SIGNIFICANCE STATEMENT Incomplete or failed myelin repairs, primarily resulting from the arrested differentiation of myelin-forming oligodendrocytes (OLs) from oligodendroglial progenitor cells, is one of the major reasons for neurologic progression in people affected by multiple sclerosis (MS). Using in vitro culture systems and in vivo animal models, this study unraveled a previously unrecognized autocrine regulation of bone morphogenetic protein (BMP)4-mediated signaling by the Wnt effector transcription factor 7-like 2 (TCF7l2). We showed for the first time that TCF7l2 promotes oligodendroglial differentiation by repressing BMP4-mediated activity, which is dysregulated in MS lesions. Our study suggests that elevating TCF7l2 expression may be possible in overcoming arrested oligodendroglial differentiation as observed in MS patients.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Regulación de la Expresión Génica/fisiología , Neurogénesis/fisiología , Oligodendroglía/citología , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Animales , Comunicación Autocrina/fisiología , Encéfalo/citología , Encéfalo/metabolismo , Diferenciación Celular/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Precursoras de Oligodendrocitos/metabolismo , Oligodendroglía/metabolismo
2.
J Neurosci ; 38(7): 1802-1820, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29335358

RESUMEN

In the CNS, myelination and remyelination depend on the successful progression and maturation of oligodendroglial lineage cells, including proliferation and differentiation of oligodendroglial progenitor cells (OPCs). Previous studies have reported that Sox2 transiently regulates oligodendrocyte (OL) differentiation in the embryonic and perinatal spinal cord and appears dispensable for myelination in the postnatal spinal cord. However, the role of Sox2 in OL development in the brain has yet to be defined. We now report that Sox2 is an essential positive regulator of developmental myelination in the postnatal murine brain of both sexes. Stage-specific paradigms of genetic disruption demonstrated that Sox2 regulated brain myelination by coordinating upstream OPC population supply and downstream OL differentiation. Transcriptomic analyses further supported a crucial role of Sox2 in brain developmental myelination. Consistently, oligodendroglial Sox2-deficient mice developed severe tremors and ataxia, typical phenotypes indicative of hypomyelination, and displayed severe impairment of motor function and prominent deficits of brain OL differentiation and myelination persisting into the later CNS developmental stages. We also found that Sox2 was required for efficient OPC proliferation and expansion and OL regeneration during remyelination in the adult brain and spinal cord. Together, our genetic evidence reveals an essential role of Sox2 in brain myelination and CNS remyelination, and suggests that manipulation of Sox2 and/or Sox2-mediated downstream pathways may be therapeutic in promoting CNS myelin repair.SIGNIFICANCE STATEMENT Promoting myelin formation and repair has translational significance in treating myelin-related neurological disorders, such as periventricular leukomalacia and multiple sclerosis in which brain developmental myelin formation and myelin repair are severely affected, respectively. In this report, analyses of a series of genetic conditional knock-out systems targeting different oligodendrocyte stages reveal a previously unappreciated role of Sox2 in coordinating upstream proliferation and downstream differentiation of oligodendroglial lineage cells in the mouse brain during developmental myelination and CNS remyelination. Our study points to the potential of manipulating Sox2 and its downstream pathways to promote oligodendrocyte regeneration and CNS myelin repair.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Encéfalo/fisiología , Sistema Nervioso Central/fisiología , Vaina de Mielina/fisiología , Oligodendroglía/fisiología , Factores de Transcripción SOXB1/fisiología , Animales , Diferenciación Celular/fisiología , Proliferación Celular , Enfermedades Desmielinizantes/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Trastornos del Movimiento/fisiopatología , Regeneración Nerviosa/fisiología , Médula Espinal/crecimiento & desarrollo , Médula Espinal/fisiología , Transcriptoma
3.
Neurobiol Dis ; 100: 30-38, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28042097

RESUMEN

Parkinson's disease (PD) is a neurodegenerative condition caused by age-related death of dopaminergic (DA) neurons in the substantia nigra (SN). Mitochondrial DNA (mtDNA) deletions rise exponentially with age in humans and reach their highest levels approaching 60% in dopaminergic neurons of the substantia nigra and overlap with dying neurons. Parkin deletion causes Parkinsonism in humans, presumably through a decrease in mitochondrial quality control, but Parkin knockout mice do not have DA neurodegeneration. We crossed Parkin knockouts to the Twinkle-TG mouse in which mtDNA deletions are increased specifically in substantia nigra to determine the effect of increased deletion mutagenesis in the absence of mitochondrial quality control. These double-mutant 'TwinkPark' mice had 1, the highest mtDNA deletion concentration in SN; 2, the lowest mitochondrial function and membrane potential; 3, the most severe neurobehavioral deficits at 19months; 4, the least dopaminergic neurons in the SN and lowest dopamine levels, i.e. Parkinsonism. This mouse model could provide novel insights into the pathomechanism by which a specific increase in mtDNA deletions with age contribute to dopaminergic neurodegeneration and Parkinson's disease.


Asunto(s)
ADN Mitocondrial/genética , Mitocondrias/genética , Trastornos Parkinsonianos/genética , Ubiquitina-Proteína Ligasas/metabolismo , Animales , ADN Mitocondrial/metabolismo , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Ratones Transgénicos , Mutación/genética , Trastornos Parkinsonianos/metabolismo , Sustancia Negra/metabolismo , Ubiquitina-Proteína Ligasas/deficiencia
4.
Hum Mol Genet ; 24(10): 2848-60, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25652399

RESUMEN

Mitochondrial complex I (NADH dehydrogenase) is a major contributor to neuronal energetics, and mutations in complex I lead to vision loss. Functional, neuroanatomical and transcriptional consequences of complex I deficiency were investigated in retinas of the Ndufs4 knockout mouse. Whole-eye ERGs and multielectrode arrays confirmed a major retinal ganglion cell functional loss at P32, and retinal ganglion cell loss at P42. RNAseq demonstrated a mild and then sharp increase in innate immune and inflammatory retinal transcripts at P22 and P33, respectively, which were confirmed with QRT-PCR. Intraperitoneal injection of the inflammogen lipopolysaccharide further reduced retinal ganglion cell function in Ndufs4 KO, supporting the connection between inflammatory activation and functional loss. Complex I deficiency in the retina clearly caused innate immune and inflammatory markers to increase coincident with loss of vision, and RGC functional loss. How complex I incites inflammation and functional loss is not clear, but could be the result of misfolded complex I generating a 'non-self' response, and induction of innate immune response transcripts was observed before functional loss at P22, including ß-2 microglobulin and Cx3cr1, and during vision loss at P31 (B2m, Tlr 2, 3, 4, C1qa, Cx3cr1 and Fas). These data support the hypothesis that mitochondrial complex I dysfunction in the retina triggers an innate immune and inflammatory response that results in loss of retinal ganglion cell function and death, as in Leber's hereditary Optic Neuropathy and suggests novel therapeutic routes to counter mitochondrial defects that contribute to vision loss.


Asunto(s)
Complejo I de Transporte de Electrón/deficiencia , Enfermedades Mitocondriales/fisiopatología , Retina/fisiopatología , Células Ganglionares de la Retina/fisiología , Animales , Muerte Celular , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/inmunología , Femenino , Técnicas de Inactivación de Genes , Inmunidad Innata/genética , Inflamación/genética , Masculino , Ratones , Ratones Noqueados , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/inmunología , Retina/inmunología
5.
Hum Mol Genet ; 21(23): 5147-58, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22949510

RESUMEN

Parkinson's disease (PD) is the second most common neurodegenerative disorder in the developed world, and is characterized by the loss of dopaminergic (DA) neurons in the substantia nigra (SN). Somatic mitochondrial DNA (mtDNA) deletions reach their highest concentration with age in the SN in humans, and may contribute to PD; yet whether mtDNA deletions cause DA neuron degeneration remains unclear. Inherited mutations of Twinkle helicase involved in mtDNA replication causes a dominant increase in mtDNA deletions in humans. We constructed a mouse model expressing mutant Twinkle in DA neurons. Mutant mice had an increase in age-related mtDNA deletions, reduction of DA neuron number in SN at 17-22 months and displayed abnormalities in rota-rod behavior. Functional analysis of midbrain indicated a slight reduction in mitochondrial state II respiration in mutants, but no decrease in maximal respiration. Also, Parkin expression was significantly decreased in DA neurons in the SN of 22-month-old mutant mice, and in PC12 cells after 48 h transfection of mutant Twinkle. Both confocal imaging and coimmunoprecipitation indicated interaction of Twinkle with Parkin in the mitochondria. Parkin overexpression rescued the reduction of proteasome activity caused by mutant Twinkle in PC12 cells. In addition, the autophagy marker LC3 was increased in the SN of 22-month transgenics, and this increase was similarly mutant Twinkle-dependent in PC12 cells. Collectively, our data demonstrate that mammalian Twinkle is important for mitochondrial integrity in DA neurons and provide a novel mouse model in which increased mtDNA deletions may lead to DA neuron degeneration and parkinsonism.


Asunto(s)
ADN Helicasas/genética , ADN Mitocondrial/genética , Neuronas Dopaminérgicas/metabolismo , Proteínas Mitocondriales/genética , Mutación , Enfermedad de Parkinson/genética , Animales , Autofagia/genética , Conducta Animal , Línea Celular , Respiración de la Célula/genética , ADN Helicasas/metabolismo , Neuronas Dopaminérgicas/patología , Expresión Génica , Orden Génico , Marcación de Gen , Humanos , Ratones , Ratones Transgénicos , Proteínas Mitocondriales/metabolismo , Enfermedad de Parkinson/metabolismo , Regiones Promotoras Genéticas , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Sustancia Negra/metabolismo , Sustancia Negra/patología , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
6.
Dev Biol ; 349(2): 250-60, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21070765

RESUMEN

The canonical Wnt/ß-catenin signaling pathway has implications in early facial development; yet, its function and signaling mechanism remain poorly understood. We report here that the frontonasal and upper jaw primordia cannot be formed after conditional ablation of ß-catenin with Foxg1-Cre mice in the facial ectoderm and the adjacent telencephalic neuroepithelium. Gene expression of several cell-survival and patterning factors, including Fgf8, Fgf3, and Fgf17, is dramatically diminished in the anterior neural ridge (ANR, a rostral signaling center) and/or the adjacent frontonasal ectoderm of the ß-catenin conditional mutant mice. In addition, Shh expression is diminished in the ventral telencephalon of the mutants, while Tcfap2a expression is less affected in the facial primordia. Apoptosis occurs robustly in the rostral head tissues following inactivation of Fgf signaling in the conditional mutants. Consequently, the upper jaw, nasal, ocular and telencephalic structures are absent, but the tongue and mandible are relatively developed in the conditional mutants at birth. Using molecular biological approaches, we demonstrate that the Fgf8 gene is transcriptionally targeted by Wnt/ß-catenin signaling during early facial and forebrain development. Furthermore, we show that conditional gain-of-function of ß-catenin signaling causes drastic upregulation of Fgf8 mRNA in the ANR and the entire facial ectoderm, which also arrests facial and forebrain development. Taken together, our results suggest that canonical Wnt/ß-catenin signaling is required for early development of the mammalian face and related head structures, which mainly or partly acts through the initiation and modulation of balanced Fgf signaling activity.


Asunto(s)
Huesos Faciales/embriología , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Transducción de Señal/fisiología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Técnicas de Ablación , Animales , Apoptosis/fisiología , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , Ectodermo/metabolismo , Técnica del Anticuerpo Fluorescente , Factores de Transcripción Forkhead/genética , Proteínas Hedgehog/metabolismo , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Luciferasas , Ratones , Ratones Mutantes , Proteínas del Tejido Nervioso/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Telencéfalo/citología , Telencéfalo/metabolismo , beta Catenina/genética
7.
Development ; 136(18): 3161-71, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19700620

RESUMEN

Neither the mechanisms that govern lip morphogenesis nor the cause of cleft lip are well understood. We report that genetic inactivation of Lrp6, a co-receptor of the Wnt/beta-catenin signaling pathway, leads to cleft lip with cleft palate. The activity of a Wnt signaling reporter is blocked in the orofacial primordia by Lrp6 deletion in mice. The morphological dynamic that is required for normal lip formation and fusion is disrupted in these mutants. The expression of the homeobox genes Msx1 and Msx2 is dramatically reduced in the mutants, which prevents the outgrowth of orofacial primordia, especially in the fusion site. We further demonstrate that Msx1 and Msx2 (but not their potential regulator Bmp4) are the downstream targets of the Wnt/beta-catenin signaling pathway during lip formation and fusion. By contrast, a ;fusion-resistant' gene, Raldh3 (also known as Aldh1a3), that encodes a retinoic acid-synthesizing enzyme is ectopically expressed in the upper lip primordia of Lrp6-deficient embryos, indicating a region-specific role of the Wnt/beta-catenin signaling pathway in repressing retinoic acid signaling. Thus, the Lrp6-mediated Wnt signaling pathway is required for lip development by orchestrating two distinctively different morphogenetic movements.


Asunto(s)
Proteínas Relacionadas con Receptor de LDL/metabolismo , Labio/embriología , Morfogénesis/fisiología , Transducción de Señal/fisiología , Proteínas Wnt/metabolismo , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Animales , Apoptosis/fisiología , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Proliferación Celular , Labio Leporino/metabolismo , Labio Leporino/patología , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas Relacionadas con Receptor de LDL/genética , Labio/anatomía & histología , Labio/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad , Factor de Transcripción MSX1/genética , Factor de Transcripción MSX1/metabolismo , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Retinal-Deshidrogenasa , Proteínas Wnt/genética
8.
Dev Dyn ; 239(1): 200-10, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19705442

RESUMEN

The role of a key Wnt coreceptor Lrp6 during heart development remains unclear. Here we show that ablation of Lrp6 in mice causes conotruncal anomalies including double-outlet right ventricle (DORV), outflow tract (OFT) cushion hypoplasia, and ventricular septal defect (VSD). Cardiac neural crest cells are specifically lost in the dorsal neural tube and caudal pharyngeal arches of the mutant embryos. We also demonstrate that Lrp6 is required for proliferation and survival of cardiac progenitors and for the expression of Isl1 in the secondary heart field. Other known cardiogenic regulators such as Msx1, Msx2, and Fgf8 are also significantly diminished in the mutant pharyngeal arches and/or OFT. Unexpectedly, the myocardium differentiation factors Mef2c and Myocardin are upregulated in the mutant OFT. Our results indicate that Lrp6 is essential for cardiac neural crest and OFT development upstream of multiple important cardiogenic genes in different cardiac lineage cells during early cardiogenesis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Cardiopatías Congénitas/genética , Corazón/embriología , Proteínas Relacionadas con Receptor de LDL/metabolismo , Cresta Neural/fisiología , Transducción de Señal/fisiología , Animales , Región Branquial/citología , Región Branquial/embriología , Bromodesoxiuridina , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Proteínas Relacionadas con Receptor de LDL/genética , Proteínas con Homeodominio LIM , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad , Factores de Transcripción MEF2 , Ratones , Ratones Mutantes , Factores Reguladores Miogénicos/metabolismo , Proteínas Nucleares/metabolismo , Transactivadores/metabolismo , Factores de Transcripción
9.
Dev Dyn ; 239(1): 318-26, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19653321

RESUMEN

Lrp6 is a key coreceptor in the canonical Wnt pathway that is widely involved in tissue/organ morphogenesis. We generated a loxP-floxed Lrp6 mouse line. Crossing with a general Cre deleter, we obtained the Lrp6-floxdel mice, in which the loxP-floxed exon 2 of Lrp6 gene has been deleted ubiquitously. The homozygotes of Lrp6-floxdel mice reproduced typical defects as seen in the conventional Lrp6-deficient mice, such as defects in eye, limb, and neural tube, and die around birth. We also found new phenotypes including cleft palate and agenesis of external genitalia in the Lrp6-floxdel mice. In addition, the Lrp6-deficient embryos are known to be defective in other systems and internal organs including the heart and brain. Thus, by selectively crossing with a lineage-specific or inducible Cre mouse line, the Lrp6 conditional gene-targeting mice will allow us to model specific types of birth defects for mechanism and prevention studies.


Asunto(s)
Anomalías Congénitas/genética , Modelos Animales de Enfermedad , Proteínas Relacionadas con Receptor de LDL/deficiencia , Animales , Northern Blotting , Cruzamientos Genéticos , Cartilla de ADN/genética , Electroporación , Exones/genética , Marcación de Gen , Immunoblotting , Proteínas Relacionadas con Receptor de LDL/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad , Ratones , Microinyecciones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Circ Res ; 101(3): 277-85, 2007 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-17585069

RESUMEN

Congenital heart diseases are the most commonly observed human birth defects and are the leading cause of infant morbidity and mortality. Accumulating evidence indicates that transforming growth factor-beta/bone morphogenetic protein signaling pathways play critical roles during cardiogenesis. Smad4 encodes the only common Smad protein in mammals, which is a critical nuclear mediator of transforming growth factor-beta/bone morphogenetic protein signaling. The aim of this work was to investigate the roles of Smad4 during heart development. To overcome the early embryonic lethality of Smad4(-/-) mice, we specifically disrupted Smad4 in the myocardium using a Cre/loxP system. We show that myocardial-specific inactivation of Smad4 caused heart failure and embryonic lethality at midgestation. Histological analysis revealed that mutant mice displayed a hypocellular myocardial wall defect, which is likely the primary cause for heart failure. Both decreased cell proliferation and increased apoptosis contributed to the myocardial wall defect in mutant mice. Data presented in this article contradict a previous report showing that Smad4 is dispensable for heart development. Our further molecular characterization showed that expression of Nmyc and its downstream targets, including cyclin D1, cyclin D2, and Id2, were downregulated in mutant embryos. Reporter analysis indicated that the transcriptional activity of the 351-bp Nmyc promoter can be positively regulated by bone morphogenetic protein stimulation and negatively regulated by transforming growth factor-beta stimulation. Chromatin immunoprecipitation analysis revealed that the Nmyc promoter can form a complex with Smad4, suggesting that Nmyc is a direct downstream target of Smad4. In conclusion, this study provides the first mouse model showing that Smad4 plays essential roles during cardiogenesis.


Asunto(s)
Corazón Fetal/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Miocardio/metabolismo , Proteína Smad4/fisiología , Secuencia de Aminoácidos , Animales , Apoptosis , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/biosíntesis , Proteínas Morfogenéticas Óseas/genética , División Celular , Femenino , Enfermedades Fetales/etiología , Genes Letales , Genes myc , Insuficiencia Cardíaca/embriología , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Miocardio/patología , Miocitos Cardíacos/patología , Células 3T3 NIH , Organogénesis , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Proteínas Proto-Oncogénicas c-myc/fisiología , Homología de Secuencia de Aminoácido , Proteína Smad4/deficiencia , Proteína Smad4/genética , Proteínas de Dominio T Box/biosíntesis , Proteínas de Dominio T Box/genética , Factor de Crecimiento Transformador beta/biosíntesis , Factor de Crecimiento Transformador beta/genética
11.
Brain Res ; 1657: 232-244, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28027875

RESUMEN

Inherited mitochondrial complex 1 deficiency causes Leber's hereditary Optic Neuropathy (LHON) and retinal ganglion cell (RGC) degeneration, and optic neuropathies are common in many inherited mitochondrial diseases. How mitochondrial defects pathomechanistically trigger optic neuropathy remains unclear. We observe that complex 1-deficient Ndufs4-/- mice present with acute vision loss around p30, and this vision loss is coincident with an 'inflammatory wave'. In order to understand what causes the inflammatory wave we explored retinal pathology that occurs from p20-p30. The results indicated that in the period p20-p30 in Ndufs4-/- retinas, there is: significant reduction in bipolar cells, RGC dendritic atrophy, reduced PSD95, increased oxidative stress as manifested by increased 4HNE, HO1 and Cuzn-SOD, increased mitochondrial biogenesis and increased apoptosis. These precede the major induction of 'inflammatory wave' at p30 shown previously, but occur earlier than frank RGC loss at p42. In general, complex 1 deficiency in retina triggers oxidative stress and mitochondrial respiratory dysfunction that causes death of the most sensitive cells, including bipolar cells and their synaptic contacts and amacrine cells in the early period, 20-24days. The early death of these cells is the likely precursor to the sharp rise in inflammatory molecules that occurs at day 30 and coincides with vision loss, and greatly precedes the death of RGCs that occurs at p42. These data suggest that metabolic antioxidant support of the most sensitive cells in the retina, or anti-inflammatory suppression of the consequences of their death, are both rational strategies for mitochondrial blinding disease.


Asunto(s)
Complejo I de Transporte de Electrón/deficiencia , Mitocondrias/metabolismo , Células Bipolares de la Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Células Amacrinas/metabolismo , Células Amacrinas/patología , Animales , Apoptosis/fisiología , Atrofia , Dendritas/metabolismo , Dendritas/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Complejo I de Transporte de Electrón/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Atrofia Óptica Hereditaria de Leber/metabolismo , Atrofia Óptica Hereditaria de Leber/patología , Biogénesis de Organelos , Estrés Oxidativo/fisiología , Densidad Postsináptica/metabolismo , Densidad Postsináptica/patología , Células Bipolares de la Retina/patología , Células Ganglionares de la Retina/patología
12.
Dalton Trans ; 46(38): 12981-12987, 2017 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-28933485

RESUMEN

A series of new lanthanide-containing peroxoisopolyoxotungstates, K6Na4[H32{Ln4(WO4)(H2O)16[W7O22(O2)2]4}3]·105H2O [Ln = CeIII (1), NdIII (2), SmIII (3), TbIII (4), ErIII (5)], have been successfully synthesized and structurally characterized. All polyanions [Ln(WO4)(H2O)16{W7O22(O2)2}4]14- are isostructural and consist of a central [Ln(WO4)(H2O)16]10+ cluster surrounded by four peripheral [W7O22(O2)2]6- units. They could act as efficient recyclable catalysts for the epoxidation of various alkenes including different cycloalkenes, styrene derivatives, internal and long-chain alkenes. Under optimal conditions, catalyst 2 displays the best catalytic activity for the oxidation of cyclooctene with high cyclooctene conversion (98.3%) and excellent selectivity (up to 99%) and could be reused for three cycles with a negligible decrease in reactivity.

13.
Cancer Lett ; 219(2): 155-62, 2005 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-15723714

RESUMEN

Although it is fairly well accepted that pulmonary tuberculosis is a major risk factor of lung cancer, the exact molecular mechanisms involved in its tumorigenesis are unclear. For this purpose, we have examined the relationship between Mycobacterium tuberculosis (M-TB) infection and FHIT gene alteration in lung cancer. Tumors with M-TB infection had a slightly higher abnormal FHIT protein expression compared with tumors without M-TB infection, although not statistically significant (Fisher's exact test, P=0.248). LOH affecting at least one locus of the FHIT gene was significantly more frequent in lung cancer patients with M-TB infection than in patients without M-TB infection whether assessment by univariate testing methods or logistic regression modeling analysis (Fisher's exact test P=0.025, logistic regression analysis P=0.012). These results indicate that M-TB infection is associated with FHIT gene LOH in lung cancer.


Asunto(s)
Ácido Anhídrido Hidrolasas/genética , Neoplasias Pulmonares/genética , Proteínas de Neoplasias/genética , Tuberculosis Pulmonar/complicaciones , Femenino , Humanos , Pérdida de Heterocigocidad , Neoplasias Pulmonares/complicaciones , Masculino , Repeticiones de Microsatélite , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Tuberculosis Pulmonar/genética
14.
Brain Res ; 1594: 82-91, 2015 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-25446449

RESUMEN

Parkinson׳s disease (PD) is the second most common neurodegenerative disorder in the developed world, and is characterized by the loss of dopaminergic (DA) neurons in the substantia nigra (SN) of midbrain. Mitochondrial complex I dysfunction has been implicated in PD pathophysiology, yet the molecular mechanism by which complex I defects may cause DA neurodegeneration remain unclear. Using Ndufs4 mouse model of mitochondrial complex I deficiency, we observed a remarkable ubiquitin protein increase in SN of Ndufs4-/- (KO) mice. By contrast, neurofilaments were significantly decreased in SN of KO mice. Furthermore, mass spectrometry and co-immunoprecipitation (Co-IP) analysis indicated an increase in ubiquitinated neurofilaments in midbrain of KO mice, whereas 20S proteasome activities were decreased, which could potentially explain the buildup of ubiquitin protein. Collectively, these data suggest that mitochondrial complex I defects cause proteasome inhibition, a consequent increase in ubiquitinated neurofilaments and other proteins, and decrease the expression of neurofilaments that could be relevant to the mechanism of DA neuronal death in PD.


Asunto(s)
Complejo I de Transporte de Electrón/metabolismo , Filamentos Intermedios/metabolismo , Enfermedad de Parkinson/metabolismo , Sustancia Negra/metabolismo , Ubiquitina/metabolismo , Animales , Western Blotting , Modelos Animales de Enfermedad , Complejo I de Transporte de Electrón/deficiencia , Inmunohistoquímica , Inmunoprecipitación , Espectrometría de Masas , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa
15.
Di Yi Jun Yi Da Xue Xue Bao ; 22(11): 992-3, 2002 Nov.
Artículo en Zh | MEDLINE | ID: mdl-12433626

RESUMEN

OBJECTIVE: To localize Mycobacterium tuberculosis DNA (TB-DNA) in lung cancer tissue and to investigate the possible relationship of this bacterial infection with the development of lung cancer. METHODS: A sensitive and specific indirect in situ nested PCR (ISNPCR) was used to identify and localize TB-DNA in 15 formalin fixed paraffin-embedded lung cancer tissue specimens, which had been demonstrated to be positive for TB-DNA by conventional PCR. RESULTS: Positive granules of TB-DNA in brown color was found mainly in the cytoplasm of the alveolar epithelial cells, pulmonary macrophages, inflammatory cells and a few tumor cells within lung cancer tissues. CONCLUSION: Mycobacterium tuberculosis infection may play a role in the pathogenesis of lung cancer.


Asunto(s)
Neoplasias Pulmonares/microbiología , Mycobacterium tuberculosis/aislamiento & purificación , Humanos , Mycobacterium tuberculosis/genética , Reacción en Cadena de la Polimerasa , Tuberculosis/etiología
16.
Cardiovasc Res ; 103(2): 258-67, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24835278

RESUMEN

AIMS: MicroRNAs (miRNAs) play critical roles during the development of the cardiovascular system. Blocking miRNA biosynthesis in embryonic hearts through a conditional gene inactivation approach led to differential cardiac defects depending on the Cre drivers used in different studies. The goal of this study is to reveal the cardiogenic pathway that is regulated by the miRNA mechanism at midgestation, a stage that has not been evaluated in previous publications. METHODS AND RESULTS: We specifically inactivated Dicer1, which is essential for generation of functional mature miRNAs, in the myocardium by crossing cTnt-Cre mice with Dicer1(loxP) mice. cTnt-Cre efficiently inactivates target genes in cardiomyocytes at midgestation. All mutants died between E14.5 and E16.5 with severe myocardial wall defects, including reduced cell proliferation, increased cell death, and spongy myocardial wall. Expression of TGFß type I receptor (Tgfbr1), which encodes the Type I receptor of TGFß ligands, was up-regulated in mutant hearts. As expected, TGFß activity was increased in Dicer1-inactivated hearts. Our further molecular analysis suggested that Tgfbr1 is a direct target of three miRNAs. Reducing TGFß activities using a pharmacological inhibitor on in vitro cultured hearts, or through an in vivo genetic approach, partially rescued the cardiac defects caused by Dicer1 inactivation. CONCLUSIONS: We show for the first time that TGFß signalling is directly regulated by the miRNA mechanism during myocardial wall morphogenesis. Increased TGFß activity plays a major role in the cardiac defects caused by myocardial deletion of Dicer1. Thus, miRNA-mediated regulation of TGFß signalling is indispensable for normal cardiogenesis.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Corazón/embriología , MicroARNs/metabolismo , Miocardio/metabolismo , Organogénesis/fisiología , Ribonucleasa III/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/fisiología
18.
Dev Dyn ; 237(11): 3157-68, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18816448

RESUMEN

Wnt reporter TOPgal mice carry a beta-galactosidase (betagal) gene under the control of the Wnt/beta-catenin signaling responsive elements. We found that the intensely immunolabeled betagal+ cells were co-immunolabeled with Nestin and formed a tangentially oriented single-cell layer in the "connecting or docking zone" where the olfactory sensory axons attached to the brain surface during mid-gestation. During early postnatal development, betagal+ cells were located in the inner olfactory nerve layer (ONLi) and co-labeled with olfactory ensheathing cell (OEC) markers S100beta and NPY but not with lineage-specific markers for neurons, oligodendrocytes, astrocytes, and microglia, demonstrating that the TOPgal marked a subpopulation of OECs. By confocal microscopy, we found that TOPgal activated processes extended along the developing glomerulus and formed multiple tunnel-like structures that ensheathe and bridge olfactory sensory axonal bundles from ONLi to the glomerulus, which may play a key role in glomerulus formation and convergent sorting of the peripheral olfactory axons.


Asunto(s)
Axones/metabolismo , Nervio Olfatorio/citología , Nervio Olfatorio/embriología , Elementos de Respuesta/fisiología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/genética , Proteínas de Filamentos Intermediarios/biosíntesis , Ratones , Ratones Transgénicos , Factores de Crecimiento Nervioso/biosíntesis , Factores de Crecimiento Nervioso/genética , Proteínas del Tejido Nervioso/biosíntesis , Nestina , Neuropéptido Y/biosíntesis , Neuropéptido Y/genética , Subunidad beta de la Proteína de Unión al Calcio S100 , Proteínas S100/biosíntesis , Proteínas S100/genética , Proteínas Wnt/genética , beta Catenina/genética
19.
Dev Dyn ; 237(12): 3681-9, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18985738

RESUMEN

Coloboma, an ocular birth defect seen in humans and other species, is caused by incomplete closure of the optic fissure. Here, we demonstrate that genetic deletion of Lrp6, a bottleneck coreceptor in the canonical Wnt signaling pathway, results in ocular coloboma and neuroretinal patterning defects in mice. The expression of ventral neuroretinal patterning gene Vax2 was conserved but with dorsally shifted expression domains; however, the dorsal neuroretinal patterning gene Tbx5 was lost in the Lrp6-mutant eyes at embryonic day 10.5. Both Bmp4 and phosphorylated Smad 1/5/8 were also significantly attenuated in the dorsal neuroretina. In addition, the retinoic acid synthesizing enzymes Raldh1 and Raldh3 were significantly changed in the mutant eyes. Our findings suggest that defective retinal patterning causes coloboma in the Lrp6-deficient mice, and that canonical Wnt signaling plays a primary role in dorsal neuroretinal patterning and related morphogenetic movements by regulation of both Bmp and retinoic acid signaling pathways.


Asunto(s)
Tipificación del Cuerpo , Coloboma/embriología , Coloboma/metabolismo , Proteínas Relacionadas con Receptor de LDL/genética , Proteínas Relacionadas con Receptor de LDL/metabolismo , Neuronas/metabolismo , Animales , Proteína Morfogenética Ósea 4/metabolismo , Coloboma/genética , Coloboma/patología , Regulación hacia Abajo , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad , Ratones , Ratones Noqueados , Mutación/genética , Transducción de Señal , Tretinoina/metabolismo , Proteínas Wnt/metabolismo
20.
Dev Biol ; 301(1): 276-86, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16959237

RESUMEN

Accumulated evidence has suggested that BMP pathways play critical roles during mammalian cardiogenesis and impairment of BMP signaling may contribute to human congenital heart diseases (CHDs), which are the leading cause of infant morbidity and mortality. Alk3 encodes a BMP specific type I receptor expressed in mouse embryonic hearts. To reveal functions of Alk3 during atrioventricular (AV) cushion morphogenesis and to overcome the early lethality of Alk3(-/-) embryos, we applied a Cre/loxp approach to specifically inactivate Alk3 in the endothelium/endocardium. Our studies showed that endocardial depletion of Alk3 severely impairs epithelium-mesenchymal-transformation (EMT) in the atrioventricular canal (AVC) region; the number of mesenchymal cells formed in Tie1-Cre;Alk3(loxp/loxp) embryos was reduced to only approximately 20% of the normal level from both in vivo section studies and in vitro explant assays. We showed, for the first time, that in addition to its functions on mesenchyme formation, Alk3 is also required for the normal growth/survival of AV cushion mesenchymal cells. Functions of Alk3 are accomplished through regulating expression/activation/subcellular localization of multiple downstream genes including Smads and cell-cycle regulators. Taken together, our study supports the notion that Alk3-mediated BMP signaling in AV endocardial/mesenchymal cells plays a central role during cushion morphogenesis.


Asunto(s)
Nodo Atrioventricular/embriología , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/fisiología , Corazón/embriología , Animales , Nodo Atrioventricular/citología , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Proliferación Celular , Supervivencia Celular , Femenino , Técnica del Anticuerpo Fluorescente , Genes Letales , Masculino , Ratones , Morfogénesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA