Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Acta Neuropathol Commun ; 11(1): 50, 2023 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-36966348

RESUMEN

Gangliogliomas are brain tumors composed of neuron-like and macroglia-like components that occur in children and young adults. Gangliogliomas are often characterized by a rare population of immature astrocyte-appearing cells expressing CD34, a marker expressed in the neuroectoderm (neural precursor cells) during embryogenesis. New insights are needed to refine tumor classification and to identify therapeutic approaches. We evaluated five gangliogliomas with single nucleus RNA-seq, cellular indexing of transcriptomes and epitopes by sequencing, and/or spatially-resolved RNA-seq. We uncovered a population of CD34+ neoplastic cells with mixed neuroectodermal, immature astrocyte, and neuronal markers. Gene regulatory network interrogation in these neuroectoderm-like cells revealed control of transcriptional programming by TCF7L2/MEIS1-PAX6 and SOX2, similar to that found during neuroectodermal/neural development. Developmental trajectory analyses place neuroectoderm-like tumor cells as precursor cells that give rise to neuron-like and macroglia-like neoplastic cells. Spatially-resolved transcriptomics revealed a neuroectoderm-like tumor cell niche with relative lack of vascular and immune cells. We used these high resolution results to deconvolute clinically-annotated transcriptomic data, confirming that CD34+ cell-associated gene programs associate with gangliogliomas compared to other glial brain tumors. Together, these deep transcriptomic approaches characterized a ganglioglioma cellular hierarchy-confirming CD34+ neuroectoderm-like tumor precursor cells, controlling transcription programs, cell signaling, and associated immune cell states. These findings may guide tumor classification, diagnosis, prognostication, and therapeutic investigations.


Asunto(s)
Neoplasias Encefálicas , Ganglioglioma , Células-Madre Neurales , Niño , Humanos , Ganglioglioma/patología , Transcriptoma , Placa Neural/patología , Células-Madre Neurales/patología , Neoplasias Encefálicas/patología
2.
Hum Mol Genet ; 29(2): 305-319, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31813957

RESUMEN

Kabuki syndrome is an autosomal dominant developmental disorder with high similarities to CHARGE syndrome. It is characterized by a typical facial gestalt in combination with short stature, intellectual disability, skeletal findings and additional features like cardiac and urogenital malformations, cleft palate, hearing loss and ophthalmological anomalies. The major cause of Kabuki syndrome are mutations in KMT2D, a gene encoding a histone H3 lysine 4 (H3K4) methyltransferase belonging to the group of chromatin modifiers. Here we provide evidence that Kabuki syndrome is a neurocrestopathy, by showing that Kmt2d loss-of-function inhibits specific steps of neural crest (NC) development. Using the Xenopus model system, we find that Kmt2d loss-of-function recapitulates major features of Kabuki syndrome including severe craniofacial malformations. A detailed marker analysis revealed defects in NC formation as well as migration. Transplantation experiments confirm that Kmt2d function is required in NC cells. Furthermore, analyzing in vivo and in vitro NC migration behavior demonstrates that Kmt2d is necessary for cell dispersion but not protrusion formation of migrating NC cells. Importantly, Kmt2d knockdown correlates with a decrease in H3K4 monomethylation and H3K27 acetylation supporting a role of Kmt2d in the transcriptional activation of target genes. Consistently, using a candidate approach, we find that Kmt2d loss-of-function inhibits Xenopus Sema3F expression, and overexpression of Sema3F can partially rescue Kmt2d loss-of-function defects. Taken together, our data reveal novel functions of Kmt2d in multiple steps of NC development and support the hypothesis that major features of Kabuki syndrome are caused by defects in NC development.


Asunto(s)
Anomalías Múltiples/enzimología , Cara/anomalías , Enfermedades Hematológicas/enzimología , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Cresta Neural/metabolismo , Enfermedades Vestibulares/enzimología , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Acetilación , Animales , Movimiento Celular/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Cara/patología , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/metabolismo , Enfermedades Hematológicas/patología , Histonas/metabolismo , Mutación con Pérdida de Función , Metilación , Mutación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Cresta Neural/enzimología , Cresta Neural/patología , Placa Neural/crecimiento & desarrollo , Placa Neural/metabolismo , Placa Neural/patología , Semaforinas/genética , Semaforinas/metabolismo , Enfermedades Vestibulares/genética , Enfermedades Vestibulares/metabolismo , Enfermedades Vestibulares/patología , Xenopus/embriología , Xenopus/genética , Xenopus/metabolismo , Proteínas de Xenopus/fisiología
3.
J Comp Pathol ; 172: 107-109, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31690407

RESUMEN

A juvenile female Moroccan uromastyx (Uromastyx acanthinurus nigriventris) that died unexpectedly was necropsied. Necropsy examination revealed minimal intracoelomic fat, small numbers of intestinal nematodes and intraocular masses within the vitreous chamber of both eyes. One of the intraocular masses was focally contiguous with the optic nerve and composed of neuroparenchyma with rare glial cells, consistent with a diagnosis of neural heterotopia. This condition is considered a neuroectodermal malformation, readily recognized in human medicine but rarely reported in animals. To the authors' knowledge, this is the first case of intraocular neural heterotopia reported in a reptile.


Asunto(s)
Coristoma/veterinaria , Lagartos , Malformaciones del Sistema Nervioso/veterinaria , Nervio Óptico/patología , Animales , Ojo/patología , Femenino , Placa Neural/crecimiento & desarrollo , Placa Neural/patología
4.
J Cell Physiol ; 234(11): 19511-19522, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30950033

RESUMEN

Mitochondrial disorders (MDs) arise as a result of a respiratory chain dysfunction. While some MDs can affect a single organ, many involve several organs, the brain being the most affected, followed by heart and/or muscle. Many of these diseases are associated with heteroplasmic mutations in the mitochondrial DNA (mtDNA). The proportion of mutated mtDNA must exceed a critical threshold to produce disease. Therefore, understanding how embryonic development determines the heteroplasmy level in each tissue could explain the organ susceptibility and the clinical heterogeneity observed in these patients. In this report, the dynamics of heteroplasmy and the influence in cardiac commitment of the mutational load of the m.13513G>A mutation has been analyzed. This mutation has been reported as a frequent cause of Leigh syndrome (LS) and is commonly associated with cardiac problems. In this report, induced pluripotent stem cell (iPSc) technology has been used to delve into the molecular mechanisms underlying cardiac disease in LS. When mutation m.13513G>A is above a threshold, iPSc-derived cardiomyocytes (iPSc-CMs) could not be obtained due to an inefficient epithelial-mesenchymal transition. Surprisingly, these cells are redirected toward neuroectodermal lineages that would give rise to the brain. However, when mutation is below that threshold, dysfunctional CM are generated in a mutant-load dependent way. We suggest that distribution of the m.13513G>A mutation during cardiac differentiation is not at random. We propose a possible explanation of why neuropathology is a frequent feature of MD, but cardiac involvement is not always present.


Asunto(s)
ADN Mitocondrial/genética , Transporte de Electrón/genética , Cardiopatías/genética , Enfermedad de Leigh/genética , Enfermedades Mitocondriales/genética , Diferenciación Celular/genética , Complejo I de Transporte de Electrón/genética , Desarrollo Embrionario/genética , Transición Epitelial-Mesenquimal/genética , Cardiopatías/patología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Enfermedad de Leigh/patología , Mitocondrias/genética , Mitocondrias/patología , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/patología , Proteínas Mitocondriales/genética , Mutación , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Placa Neural/crecimiento & desarrollo , Placa Neural/patología , Fenotipo
5.
Development ; 145(9)2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29636380

RESUMEN

The last stage of neural tube (NT) formation involves closure of the caudal neural plate (NP), an embryonic structure formed by neuromesodermal progenitors and newly differentiated cells that becomes incorporated into the NT. Here, we show in mouse that, as cell specification progresses, neuromesodermal progenitors and their progeny undergo significant changes in shape prior to their incorporation into the NT. The caudo-rostral progression towards differentiation is coupled to a gradual reliance on a unique combination of complex mechanisms that drive tissue folding, involving pulses of apical actomyosin contraction and planar polarised cell rearrangements, all of which are regulated by the Wnt-PCP pathway. Indeed, when this pathway is disrupted, either chemically or genetically, the polarisation and morphology of cells within the entire caudal NP is disturbed, producing delays in NT closure. The most severe disruptions of this pathway prevent caudal NT closure and result in spina bifida. In addition, a decrease in Vangl2 gene dosage also appears to promote more rapid progression towards a neural fate, but not the specification of more neural cells.


Asunto(s)
Diferenciación Celular , Placa Neural/embriología , Células-Madre Neurales/metabolismo , Tubo Neural/embriología , Vía de Señalización Wnt , Animales , Ratones , Ratones Mutantes , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Placa Neural/patología , Células-Madre Neurales/patología , Tubo Neural/patología , Disrafia Espinal/epidemiología , Disrafia Espinal/genética , Disrafia Espinal/patología
6.
PLoS One ; 11(12): e0167573, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27907123

RESUMEN

Charcot-Marie-Tooth disease type 2A (CMT2A), the most common axonal form of hereditary sensory motor neuropathy, is caused by mutations of mitofusin-2 (MFN2). Mitofusin-2 is a GTPase required for fusion of mitochondrial outer membranes, repair of damaged mitochondria, efficient mitochondrial energetics, regulation of mitochondrial-endoplasmic reticulum calcium coupling and axonal transport of mitochondria. We knocked T105M MFN2 preceded by a loxP-flanked STOP sequence into the mouse Rosa26 locus to permit cell type-specific expression of this pathogenic allele. Crossing these mice with nestin-Cre transgenic mice elicited T105M MFN2 expression in neuroectoderm, and resulted in diminished numbers of mitochondria in peripheral nerve axons, an alteration in skeletal muscle fiber type distribution, and a gait abnormality.


Asunto(s)
Alelos , Enfermedad de Charcot-Marie-Tooth/genética , GTP Fosfohidrolasas/genética , Hemicigoto , Fibras Musculares Esqueléticas/metabolismo , Nervios Periféricos/metabolismo , Sustitución de Aminoácidos , Animales , Transporte Axonal , Enfermedad de Charcot-Marie-Tooth/metabolismo , Enfermedad de Charcot-Marie-Tooth/patología , Modelos Animales de Enfermedad , Embrión de Mamíferos , GTP Fosfohidrolasas/deficiencia , Marcha , Regulación de la Expresión Génica , Miembro Posterior/patología , Humanos , Integrasas/genética , Integrasas/metabolismo , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Mitocondrias/patología , Fibras Musculares Esqueléticas/patología , Nestina/genética , Nestina/metabolismo , Placa Neural/metabolismo , Placa Neural/patología , Nervios Periféricos/patología , Fenotipo , ARN no Traducido/genética , ARN no Traducido/metabolismo
7.
Dev Biol ; 415(2): 371-382, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-26874011

RESUMEN

Mandibulofacial dysostosis (MFD) is a human developmental disorder characterized by defects of the facial bones. It is the second most frequent craniofacial malformation after cleft lip and palate. Nager syndrome combines many features of MFD with a variety of limb defects. Mutations in SF3B4 (splicing factor 3b, subunit 4) gene, which encodes a component of the pre-mRNA spliceosomal complex, were recently identified as a cause of Nager syndrome, accounting for 60% of affected individuals. Nothing is known about the cellular pathogenesis underlying Nager type MFD. Here we describe the first animal model for Nager syndrome, generated by knocking down Sf3b4 function in Xenopus laevis embryos, using morpholino antisense oligonucleotides. Our results indicate that Sf3b4-depleted embryos show reduced expression of the neural crest genes sox10, snail2 and twist at the neural plate border, associated with a broadening of the neural plate. This phenotype can be rescued by injection of wild-type human SF3B4 mRNA but not by mRNAs carrying mutations that cause Nager syndrome. At the tailbud stage, morphant embryos had decreased sox10 and tfap2a expression in the pharyngeal arches, indicative of a reduced number of neural crest cells. Later in development, Sf3b4-depleted tadpoles exhibited hypoplasia of neural crest-derived craniofacial cartilages, phenocopying aspects of the craniofacial skeletal defects seen in Nager syndrome patients. With this animal model we are now poised to gain important insights into the etiology and pathogenesis of Nager type MFD, and to identify the molecular targets of Sf3b4.


Asunto(s)
Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica/genética , Disostosis Mandibulofacial/genética , Desarrollo Maxilofacial/genética , Factores de Empalme de ARN/genética , Proteínas de Xenopus/deficiencia , Xenopus laevis/genética , Secuencia de Aminoácidos , Animales , Región Branquial/embriología , Región Branquial/metabolismo , Región Branquial/patología , Cartílago/crecimiento & desarrollo , Cartílago/metabolismo , Cartílago/patología , Codón sin Sentido , Mutación del Sistema de Lectura , Técnicas de Silenciamiento del Gen , Prueba de Complementación Genética , Humanos , Disostosis Mandibulofacial/embriología , Disostosis Mandibulofacial/fisiopatología , Datos de Secuencia Molecular , Morfolinos/farmacología , Cresta Neural/citología , Cresta Neural/embriología , Cresta Neural/metabolismo , Placa Neural/embriología , Placa Neural/patología , Fenotipo , Precursores del ARN/genética , Precursores del ARN/metabolismo , Empalme del ARN/genética , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Cráneo/anomalías , Cráneo/embriología , Cráneo/crecimiento & desarrollo , Proteínas de Xenopus/biosíntesis , Proteínas de Xenopus/genética , Proteínas de Xenopus/fisiología , Xenopus laevis/embriología , Xenopus laevis/crecimiento & desarrollo
8.
Sci Rep ; 5: 16917, 2015 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-26593875

RESUMEN

Failure to close the neural tube results in birth defects, with severity ranging from spina bifida to lethal anencephaly. Few genetic risk factors for neural tube defects are known in humans, highlighting the critical role of environmental risk factors, such as maternal diabetes. Yet, it is not well understood how altered maternal metabolism interferes with embryonic development, and with neurulation in particular. We present evidence from two independent mouse models of diabetic pregnancy that identifies impaired migration of nascent mesodermal cells in the primitive streak as the morphogenetic basis underlying the pathogenesis of neural tube defects. We conclude that perturbed gastrulation not only explains the neurulation defects, but also provides a unifying etiology for the broad spectrum of congenital malformations in diabetic pregnancies.


Asunto(s)
Diabetes Gestacional/genética , Proteínas del Tejido Nervioso/genética , Placa Neural/metabolismo , Defectos del Tubo Neural/genética , Animales , Diabetes Gestacional/metabolismo , Diabetes Gestacional/patología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Femenino , Gastrulación/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Captura por Microdisección con Láser , Ratones , Ratones Endogámicos NOD , Proteínas del Tejido Nervioso/metabolismo , Placa Neural/embriología , Placa Neural/patología , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/patología , Embarazo
9.
Mol Brain ; 7: 67, 2014 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-25223405

RESUMEN

BACKGROUND: Alcohol is detrimental to early development. Fetal alcohol spectrum disorders (FASD) due to maternal alcohol abuse results in a series of developmental abnormalities including cranial facial dysmorphology, ocular anomalies, congenital heart defects, microcephaly and intellectual disabilities. Previous studies have been shown that ethanol exposure causes neural crest (NC) apoptosis and perturbation of neural crest migration. However, the underlying mechanism remains elusive. In this report we investigated the fetal effect of alcohol on the process of neural crest development in the Xenopus leavis. RESULTS: Pre-gastrulation exposure of 2-4% alcohol induces apoptosis in Xenopus embryo whereas 1% alcohol specifically impairs neural crest migration without observing discernible apoptosis. Additionally, 1% alcohol treatment considerably increased the phenotype of small head (43.4% ± 4.4%, total embryo n = 234), and 1.5% and 2.0% dramatically augment the deformation to 81.2% ± 6.5% (n = 205) and 91.6% ± 3.0% (n = 235), respectively (P < 0.05). Significant accumulation of Homocysteine was caused by alcohol treatment in embryos and 5-mehtyltetrahydrofolate restores neural crest migration and alleviates homocysteine accumulation, resulting in inhibition of the alcohol-induced neurocristopathies. CONCLUSIONS: Our study demonstrates that prenatal alcohol exposure causes neural crest cell migration abnormality and 5-mehtyltetrahydrofolate could be beneficial for treating FASD.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Etanol/toxicidad , Cresta Neural/patología , Tetrahidrofolatos/farmacología , Animales , Apoptosis/efectos de los fármacos , Cartílago/efectos de los fármacos , Cartílago/embriología , Modelos Animales , Cresta Neural/efectos de los fármacos , Placa Neural/efectos de los fármacos , Placa Neural/patología , Pigmentos Biológicos/metabolismo , Xenopus laevis/embriología
11.
Dev Med Child Neurol ; 56(12): 1207-1211, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24954233

RESUMEN

AIM: Patients with tuberous sclerosis complex (TSC) with brain involvement usually have both tubers and subependymal nodules (SENs) and the occurrence of one lesion without the other seems to be rare. The aim of this study was to assess the specific clinical manifestations and genotype of patients with one type of lesion or the other but not both. METHOD: The magnetic resonance images of 220 patients with TSC were reviewed, and patients with either tubers or SENs, but not both, were identified. RESULTS: Out of a total of 220 patients (95 males, 125 females; mean age 22y 9mo, range 9mo-81y), six (3%) had tubers without SENs (two males; four females; mean age 34y 10mo, range 11-48y); however, no patients with SENs and without tubers were identified. No mutation was identified (NMI) in any of the six patients who underwent mutational analysis of TSC1 and TSC2. Five of the six patients had three or fewer tubers. INTERPRETATION: We found no patients with SENs but without tubers among our cohort. In all patients with tubers but without SENs, mutational studies of TSC1/TSC2 were negative, and the majority of these had three or fewer tubers. A possible mechanism for patients with NMI and an absence of SENs is a mosaicism with a first postzygotic mutation in the neuroectoderm.


Asunto(s)
Encéfalo/patología , Mosaicismo , Placa Neural/patología , Esclerosis Tuberosa/genética , Esclerosis Tuberosa/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Femenino , Humanos , Lactante , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Mutación/genética , Estudios Retrospectivos , Adulto Joven
12.
Reprod Toxicol ; 48: 88-97, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24887031

RESUMEN

Malformations of the facial midline are a consistent feature among individuals with defects in primary cilia. Here, we provide a framework in which to consider how these primary cilia-dependent facial anomalies occur. We generated mice in which the intraflagellar transport protein Kif3a was deleted in cranial neural crest cells. The Kif3a phenotypes included isolated metopic craniosynostosis, delayed closure of the anterior fontanelles, and hydrocephalus, as well as midline facial anomalies including hypertelorism, cleft palate, and bifid nasal septum. Although all cranial neural crest cells had truncated primary cilia as a result of the conditional deletion, only those in the midline showed evidence of hyper-proliferation and ectopic Wnt responsiveness. Thus, cranial neural crest cells do not rely on primary cilia for their migration but once established in the facial prominences, midline cranial neural crest cells require Kif3a function in order to integrate and respond to Wnt signals from the surrounding epithelia.


Asunto(s)
Cilios/fisiología , Fisura del Paladar/genética , Craneosinostosis/genética , Hidrocefalia/genética , Hipertelorismo/genética , Cinesinas/genética , Animales , Fontanelas Craneales/anomalías , Ratones Transgénicos , Mutación , Cresta Neural/metabolismo , Cresta Neural/patología , Placa Neural/metabolismo , Placa Neural/patología , Fenotipo , Vía de Señalización Wnt
13.
Artículo en Inglés | MEDLINE | ID: mdl-24902834

RESUMEN

The neural tube (NT), the embryonic precursor of the vertebrate brain and spinal cord, is generated by a complex and highly dynamic morphological process. In mammals, the initially flat neural plate bends and lifts bilaterally to generate the neural folds followed by fusion of the folds at the midline during the process of neural tube closure (NTC). Failures in any step of this process can lead to neural tube defects (NTDs), a common class of birth defects that occur in approximately 1 in 1000 live births. These severe birth abnormalities include spina bifida, a failure of closure at the spinal level; craniorachischisis, a failure of NTC along the entire body axis; and exencephaly, a failure of the cranial neural folds to close which leads to degeneration of the exposed brain tissue termed anencephaly. The mouse embryo presents excellent opportunities to explore the genetic basis of NTC in mammals; however, its in utero development has also presented great challenges in generating a deeper understanding of how gene function regulates the cell and tissue behaviors that drive this highly dynamic process. Recent technological advances are now allowing researchers to address these questions through visualization of NTC dynamics in the mouse embryo in real time, thus offering new insights into the morphogenesis of mammalian NTC.


Asunto(s)
Morfogénesis , Cresta Neural/crecimiento & desarrollo , Placa Neural/crecimiento & desarrollo , Tubo Neural/crecimiento & desarrollo , Anencefalia/genética , Anencefalia/patología , Animales , Ratones , Placa Neural/patología , Tubo Neural/patología , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/patología , Disrafia Espinal/genética , Disrafia Espinal/patología
14.
Cell Stem Cell ; 14(1): 27-39, 2014 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-24268696

RESUMEN

Although somatic cell nuclear transfer (SCNT) and induction of pluripotency (to form iPSCs) are both recognized reprogramming methods, there has been relatively little comparative analysis of the resulting pluripotent cells. Here, we examine the capacity of these two reprogramming approaches to rejuvenate telomeres using late-generation telomerase-deficient (Terc(-/-)) mice that exhibit telomere dysfunction and premature aging. We found that embryonic stem cells established from Terc(-/-) SCNT embryos (Terc(-/-) ntESCs) have greater differentiation potential and self-renewal capacity than Terc(-/-) iPSCs. Remarkably, SCNT results in extensive telomere lengthening in cloned embryos and improved telomere capping function in the established Terc(-/-) ntESCs. In addition, mitochondrial function is severely impaired in Terc(-/-) iPSCs and their differentiated derivatives but significantly improved in Terc(-/-) ntESCs. Thus, our results suggest that SCNT-mediated reprogramming mitigates telomere dysfunction and mitochondrial defects to a greater extent than iPSC-based reprogramming. Understanding the basis of this differential could help optimize reprogramming strategies.


Asunto(s)
Diferenciación Celular , Reprogramación Celular , Células Madre Embrionarias/citología , Células Madre Pluripotentes Inducidas/citología , Técnicas de Transferencia Nuclear , ARN/fisiología , Telomerasa/fisiología , Telómero/genética , Adenosina Trifosfato/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Células Madre Embrionarias/metabolismo , Hibridación Fluorescente in Situ , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/patología , Placa Neural/metabolismo , Placa Neural/patología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Curr Med Chem ; 21(8): 1017-25, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23992320

RESUMEN

Advanced enteropancreatic (EP) neuroendocrine tumors (NETs) can be treated with several different therapies, including chemotherapy, biotherapy, and locoregional treatments. Over the last few decades, impressive progress has been made in the biotherapy field. Three main druggable molecular targets have been studied and developed in terms of therapy: somatostatin receptor (sstr), mammalian target of rapamycin (mTOR), and angiogenic factors. In particular, research has moved from the old somatostatin analogs (SSAs), such as octreotide (OCT) and lanreotide (LAN), specifically binding to the sstr-2, to the newer pasireotide (PAS), which presents a wider sstr spectrum. Over the last ten years, several molecular targeted agents (MTAs) have been studied in phase II trials, and very few of them have reached phase III. The mTOR inhibitor everolimus and the multitargeted inhibitor sunitinib have been approved for clinical use by the FDA and EMA in advanced well/moderately-differentiated (WD, MD) progressive pancreatic neuroendocrine tumors (PNETs), on the basis of the positive results of two international large randomized phase III trials vs. placebo. Bevacizumab has been studied in a large US phase III trial vs. interferon (IFN)-alfa2b, and results are pending. In this review, the biological and clinical aspects of MTAs introduced into clinical practice or which are currently in an advanced phase of clinical investigation are addressed.


Asunto(s)
Antineoplásicos/uso terapéutico , Terapia Molecular Dirigida/métodos , Placa Neural/patología , Tumores Neuroendocrinos/tratamiento farmacológico , Páncreas/patología , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Humanos , Placa Neural/efectos de los fármacos , Placa Neural/metabolismo , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología
16.
J Oral Pathol Med ; 42(6): 450-3, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23336292

RESUMEN

BACKGROUND: The glucose transporter type 1 (GLUT-1) protein is a useful marker for perineurial cells. Because of the possible neuroectodermal histogenesis of the granular cell tumour and congenital granular cell epulis, the aim of this study was to assess the immunoexpression of GLUT-1 protein in granular cell tumour and congenital granular cell epulis to aid in clarifying their histogenesis. METHODS: The protocol of this study was approved by the Committee of Bioethics in Research at Universidade Federal Minas Gerais. Six cases of granular cell tumour and three cases of congenital granular cell epulis were submitted to immunohistochemistry for GLUT-1 and S-100 using the streptavidin-biotin standard protocol. RESULTS: Five cases of granular cell tumour were located on the tongue and one case on the upper lip. All cases of congenital granular cell epulis were observed in the alveolar ridge of newborns. All lesions evaluated proved to be immunonegative for GLUT-1. S-100 was found to be positive in all granular cell tumours and negative in congenital granular cell epulis. CONCLUSIONS: Neither granular cell tumour nor congenital granular cell epulis is directly related to perineurial cells.


Asunto(s)
Neoplasias Gingivales/congénito , Transportador de Glucosa de Tipo 1/análisis , Tumor de Células Granulares/patología , Adulto , Femenino , Neoplasias Gingivales/patología , Humanos , Inmunohistoquímica , Recién Nacido , Neoplasias de los Labios/patología , Masculino , Persona de Mediana Edad , Placa Neural/patología , Nervios Periféricos/patología , Proteínas S100/análisis , Neoplasias de la Lengua/patología
17.
Hum Mol Genet ; 21(18): 4104-14, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22723015

RESUMEN

Exposure to the antiepileptic drug valproic acid (VPA) during gestation causes neurofunctional and anatomic deficits in later life. At present, there are little human data on how early neural development is affected by chemicals. We used human embryonic stem cells, differentiating to neuroectodermal precursors, as a model to investigate the modes of action of VPA. Microarray expression profiling, qPCR of specific marker genes, immunostaining and the expression of green fluorescent protein under the control of the promoter of the canonical neural precursor cell marker HES5 were used as readouts. Exposure to VPA resulted in distorted marker gene expression, characterized by a relative increase in NANOG and OCT4 and a reduction in PAX6. A similar response pattern was observed with trichostatin A, a potent and specific histone deacetylase inhibitor (HDACi), but not with several other toxicants. Differentiation markers were disturbed by prolonged, but not by acute treatment with HDACi, and the strongest disturbance of differentiation was observed by toxicant exposure during early neural fate decision. The increased acetylation of histones observed in the presence of HDACi may explain the up-regulation of some genes. However, to understand the down-regulation of PAX6 and the overall complex transcript changes, we examined further epigenetic markers. Alterations in the methylation of lysines 4 and 27 of histone H3 were detected in the promoter region of PAX6 and OCT4. The changes in these activating and silencing histone marks provide a more general mechanistic rational for the regulation of developmentally important genes at non-cytotoxic drug concentrations.


Asunto(s)
Anomalías Inducidas por Medicamentos/genética , Células Madre Embrionarias/metabolismo , Epigénesis Genética/efectos de los fármacos , Placa Neural/embriología , Anomalías Inducidas por Medicamentos/patología , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Células Madre Embrionarias/fisiología , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Metilación , Proteína Homeótica Nanog , Placa Neural/patología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/fisiología , Células Neuroepiteliales/metabolismo , Células Neuroepiteliales/fisiología , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Análisis de Componente Principal , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transcripción Genética , Transcriptoma , Ácido Valproico/efectos adversos
18.
Cytotherapy ; 12(4): 491-504, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20331410

RESUMEN

BACKGROUND AIMS: In recent years, bone marrow (BM)-derived mesenchymal stromal cells (MSC) have become a promising source for neuroregenerative therapies. We evaluated the trophic effects of neuroectodermally converted MSC (mNSC) on neural stem cells (NSC). METHODS: We quantified the expression of growth factors by mNSC using real-time reverse transcription-polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA) and studied the effects of mNSC conditioned medium and mNSC (in direct co-culture) on NSC proliferation, differentiation and survival. RESULTS: Neuroectodermal conversion of human MSC induced high expression of growth factors at both mRNA and protein levels, most prominently hepatocyte growth factor, vascular endothelial growth factor and amphiregulin (37 +/- 17, 92 +/- 44 and 12 +/- 11 ng/10(5) cells, respectively), which remained at high levels upon co-culturing with neural cells. Accordingly, mNSC conditioned medium and co-cultivation with mNSC reduced cell death of NSC (36% of control), stimulated their proliferation, attenuated glial differentiation of NSC (7 +/- 3 versus 59 +/- 6%; P < 0.01) and protected NSC against the neurotoxin 6-hydroxydopamine (with half-maximally concentrations EC(50) values of 217 +/- 207 microM in the presence of mNSC compared with 62 +/- 49 microM for NSC alone). CONCLUSIONS: mNSC promote survival and proliferation, and inhibit glial differentiation, of NSC. Protection of NSC by mNSC against 6-hydroxy-dopamine is probably mediated by the release of cytotrophic factors. Our results promote neuroectodermally converted MSC as promising candidate cells for the development of neuroregenerative and neuroprotective therapies.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas/metabolismo , Placa Neural/metabolismo , Neuronas/metabolismo , Células del Estroma/metabolismo , Adolescente , Adulto , Animales , Apoptosis/efectos de los fármacos , Comunicación Celular , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular , Proliferación Celular/efectos de los fármacos , Técnicas de Cocultivo , Medios de Cultivo Condicionados/metabolismo , Medios de Cultivo Condicionados/farmacología , Citoprotección/genética , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/patología , Ratones , Persona de Mediana Edad , Placa Neural/patología , Neuroglía/metabolismo , Neuroglía/patología , Neuronas/efectos de los fármacos , Neuronas/patología , Oxidopamina/toxicidad , Ratas , Células del Estroma/efectos de los fármacos , Células del Estroma/patología
19.
Proc Natl Acad Sci U S A ; 106(13): 5324-9, 2009 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-19289832

RESUMEN

Ewing tumors (ET) are highly malignant, localized in bone or soft tissue, and are molecularly defined by ews/ets translocations. DNA microarray analysis revealed a relationship of ET to both endothelium and fetal neural crest. We identified expression of histone methyltransferase enhancer of Zeste, Drosophila, Homolog 2 (EZH2) to be increased in ET. Suppressive activity of EZH2 maintains stemness in normal and malignant cells. Here, we found EWS/FLI1 bound to the EZH2 promoter in vivo, and induced EZH2 expression in ET and mesenchymal stem cells. Down-regulation of EZH2 by RNA interference in ET suppressed oncogenic transformation by inhibiting clonogenicity in vitro. Similarly, tumor development and metastasis was suppressed in immunodeficient Rag2(-/-)gamma(C)(-/-) mice. EZH2-mediated gene silencing was shown to be dependent on histone deacetylase (HDAC) activity. Subsequent microarray analysis of EZH2 knock down, HDAC-inhibitor treatment and confirmation in independent assays revealed an undifferentiated phenotype maintained by EZH2 in ET. EZH2 regulated stemness genes such as nerve growth factor receptor (NGFR), as well as genes involved in neuroectodermal and endothelial differentiation (EMP1, EPHB2, GFAP, and GAP43). These data suggest that EZH2 might have a central role in ET pathology by shaping the oncogenicity and stem cell phenotype of this tumor.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Células Endoteliales/patología , Placa Neural/patología , Sarcoma de Ewing/etiología , Factores de Transcripción/fisiología , Animales , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Proteína Potenciadora del Homólogo Zeste 2 , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Histona Desacetilasas , Humanos , Células Madre Mesenquimatosas , Ratones , Metástasis de la Neoplasia , Proteínas de Fusión Oncogénica , Complejo Represivo Polycomb 2 , Proteína Proto-Oncogénica c-fli-1 , Proteína EWS de Unión a ARN , Sarcoma de Ewing/patología
20.
Diagn Cytopathol ; 36(8): 595-9, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18618728

RESUMEN

Immature ovarian teratoma (IOT) is a rare and aggressive malignant neoplasm characterized by immature neural tissue. The cytomorphologic features have only rarely been described. We herein describe an additional case and review the literature regarding this entity. To the best of our knowledge, this is the first reported case with imprint cytology. A 35-year-old woman presented with a pelvic mass which was resected and sent for frozen section evaluation. Imprint smears and frozen section of the mass were diagnostic of IOT. IOT has diagnostic cytologic features which show complete concordance with histology. Differential diagnoses include other small round cell neoplasms such as ovarian neuroblastoma, small cell carcinoma of hypercalcemic type, primitive neuroectodermal tumor, Wilm's tumor, desmoplastic small round cell tumor, and Non-Hodgkin lymphoma. Distinguishing IOT from these tumors can be challenging however if diligent morphologic study and/or ancillary studies are performed accurate diagnosis is possible.


Asunto(s)
Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/patología , Teratoma/diagnóstico , Teratoma/patología , Adulto , Diagnóstico Diferencial , Femenino , Humanos , Placa Neural/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...