Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Commun Med (Lond) ; 3(1): 177, 2023 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-38082066

RESUMEN

BACKGROUND: Pregnant women and their fetuses are particularly susceptible to respiratory pathogens. How they respond to SARS-CoV-2 infection is still under investigation. METHODS: We studied the transcriptome and phenotype of umbilical cord blood cells in pregnant women infected or not with SARS-CoV-2. RESULTS: Here we show that symptomatic maternal COVID-19 is associated with a transcriptional erythroid cell signature as compared with asymptomatic and uninfected mothers. We observe an expansion of fetal hematopoietic multipotent progenitors skewed towards erythroid differentiation that display increased clonogenicity. There was no difference in inflammatory cytokines levels in the cord blood upon maternal SARS-CoV-2 infection. Interestingly, we show an activation of hypoxia pathway in cord blood cells from symptomatic COVID-19 mothers, suggesting that maternal hypoxia may be triggering this fetal stress hematopoiesis. CONCLUSIONS: Overall, these results show a fetal hematopoietic response to symptomatic COVID-19 in pregnant mothers in the absence of vertically transmitted SARS-CoV-2 infection which is likely to be a mechanism of fetal adaptation to the maternal infection and reduced oxygen supply.


During pregnancy, women are more prone to respiratory infectious diseases. It is not known if COVID-19 infection has an adverse effect on the growing fetus. Here, we aimed to identify any potential effects of COVID-19 infection on the fetus by taking measurements from the umbilical cord blood cells. In mothers who displayed symptomatic COVID-19 infection, we observed an increased production of hematopoietic progenitor cells, especially the ones that are responsible for producing red blood cells. We think this might be a coping mechanism for the fetus, as the mother's body deals with the infection. Therefore, our work shows that growing fetuses do respond to maternal COVID-19 symptoms, even when they are protected in the womb from the infection and may never get infected by the mother.

2.
Aging (Albany NY) ; 14(9): 3728-3756, 2022 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-35507806

RESUMEN

Naked mole-rats (NMR) are subterranean rodents characterized by an unusual longevity coupled with an unexplained resistance to aging. In the present study, we performed extensive in situ analysis and single-cell RNA-sequencing comparing young and older animals. At variance with other species, NMR exhibited a striking stability of skin compartments and cell types, which remained stable over time without aging-associated changes. Remarkably, the number of stem cells was constant throughout aging. We found three classical cellular states defining a unique keratinocyte differentiation trajectory that were not altered after pseudo-temporal reconstruction. Epidermal gene expression did not change with aging either. Langerhans cell clusters were conserved, and only a higher basal stem cell expression of Igfbp3 was found in aged animals. In accordance, NMR skin healing closure was similar in young and older animals. Altogether, these results indicate that NMR skin is characterized by peculiar genetic and cellular features, different from those previously demonstrated for mice and humans. The remarkable stability of the aging NMR skin transcriptome likely reflects unaltered homeostasis and resilience.


Asunto(s)
Ratas Topo , Transcriptoma , Envejecimiento/genética , Animales , Longevidad/genética , Ratones , Ratas Topo/genética , Células Madre
3.
J Invest Dermatol ; 139(9): 2004-2015.e13, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31059696

RESUMEN

The management of large congenital melanocytic nevi (lCMN) is based exclusively on iterative surgical procedures in the absence of validated medical therapy. The aim of our study was to develop an intra-lesional medical treatment for lCMN. Seventeen patients harboring NRAS-mutated lCMN were included. Nevocytes obtained from lCMN displayed an overactivation of mitogen-activated protein kinase and phosphoinositide 3-kinase (Akt) pathways. Mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK) and Akt inhibitors reduced the nevosphere diameter in sphere-forming assays, as well as cell viability and proliferation in in vitro assays. Standardized lCMN explants were then cultured ex vivo with the same inhibitors, which induced a decrease in MelanA+ and Sox10+ cells in both epidermis and dermis. Finally, intradermal injections of these inhibitors were administered within standardized lCMN xenografts in Rag2-/- mice. They induced a dramatic decrease in nevocytes in treated xenografts, which persisted 30 days after the end of treatment. Using original nevus explant and xenograft preclinical models, we demonstrated that intradermal MEK/Akt inhibition might serve as neoadjuvant therapy for the treatment of NRAS-mutated congenital melanocytic nevi to avoid iterative surgeries.


Asunto(s)
Antineoplásicos/administración & dosificación , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Nevo Pigmentado/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Proliferación Celular/efectos de los fármacos , Niño , Preescolar , Femenino , GTP Fosfohidrolasas/genética , Humanos , Lactante , Inyecciones Intradérmicas , Inyecciones Intralesiones , Antígeno MART-1/metabolismo , Masculino , Melanocitos/efectos de los fármacos , Melanocitos/patología , Proteínas de la Membrana/genética , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Nevo Pigmentado/congénito , Nevo Pigmentado/genética , Nevo Pigmentado/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción SOXE/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Piel/citología , Piel/patología , Neoplasias Cutáneas/congénito , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Transl Med ; 16(1): 131, 2018 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-29783999

RESUMEN

BACKGROUND: Actual European pathological classification of early-stage endometrial cancer (EC) may show insufficient accuracy to precisely stratify recurrence risk, leading to potential over or under treatment. Micro-RNAs are post-transcriptional regulators involved in carcinogenic mechanisms, with some micro-RNA patterns of expression associated with EC characteristics and prognosis. We previously demonstrated that downregulation of micro-RNA-184 was associated with lymph node involvement in low-risk EC (LREC). The aim of this study was to evaluate whether micro-RNA signature in tumor tissues from LREC women can be correlated with the occurrence of recurrences. METHODS: MicroRNA expression was assessed by chip analysis and qRT-PCR in 7 formalin-fixed paraffin-embedded (FFPE) LREC primary tumors from women whose follow up showed recurrences (R+) and in 14 FFPE LREC primary tumors from women whose follow up did not show any recurrence (R-), matched for grade and age. Various statistical analyses, including enrichment analysis and a minimum p-value approach, were performed. RESULTS: The expression levels of micro-RNAs-184, -497-5p, and -196b-3p were significantly lower in R+ compared to R- women. Women with a micro-RNA-184 fold change < 0.083 were more likely to show recurrence (n = 6; 66%) compared to those with a micro-RNA-184 fold change > 0.083 (n = 1; 8%), p = 0.016. Women with a micro-RNA-196 fold change < 0.56 were more likely to show recurrence (n = 5; 100%) compared to those with a micro-RNA-196 fold change > 0.56 (n = 2; 13%), p = 0.001. CONCLUSIONS: These findings confirm the great interest of micro-RNA-184 as a prognostic tool to improve the management of LREC women.


Asunto(s)
Neoplasias Endometriales/genética , Perfilación de la Expresión Génica , MicroARNs/genética , Recurrencia Local de Neoplasia/genética , Anciano , Anciano de 80 o más Años , Regulación hacia Abajo/genética , Neoplasias Endometriales/epidemiología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/metabolismo , Persona de Mediana Edad , Factores de Riesgo , Regulación hacia Arriba/genética
5.
Exp Dermatol ; 25(10): 789-96, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27193390

RESUMEN

Congenital melanocytic nevi (CMN) are benign proliferations that may be associated with various consequences depending on their size. They are characterized by a specific molecular signature, namely a postzygotic somatic NRAS or BRAF mutation. We have recently reported that large CMN (lCMN), which are classically associated with an increased melanoma risk, harbour cell subpopulations with specific clonogenic and tumorigenic potential. We wished to ascertain whether cells displaying similar properties persisted postnatally in medium CMN (mCMN). Eighteen medium M1, nine large and one giant NRAS-mutated CMN were prospectively included in the study. Subpopulations of mCMN cells expressed stem cell/progenitor lineage markers such as Sox10, nestin and Oct4, as was the case in lCMN. Nevertheless, conversely to lCMN, mCMN cells with clonogenic properties were rarer. In vitro, approximatively one in 1500 cells isolated from fresh mCMN formed colonies that could be passaged. In vivo, mCMN seemed to harbour cells with less proliferative potential than the larger lesions as lCMN biopsies displayed a threefold expansion compared to mCMN when xenografted in Rag2(-/-) mice. Thus, our data revealed variations in clonogenicity and tumorigenic properties in NRAS-mutated CMN according to size.


Asunto(s)
Carcinogénesis , Melanoma/etiología , Nevo Pigmentado/patología , Piel/patología , Adolescente , Biomarcadores/metabolismo , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Nevo Pigmentado/congénito , Nevo Pigmentado/genética , Nevo Pigmentado/metabolismo , Células Madre/metabolismo
6.
J Invest Dermatol ; 135(3): 824-833, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25310409

RESUMEN

Large congenital melanocytic nevi (lCMN) are benign melanocytic tumors associated with an increased risk of melanoma transformation. They result predominantly from a post-zygotic somatic NRAS mutation. These lesions persist and even increase after birth proportionally to the child's growth. Therefore, we asked here whether cells with clonogenic and tumorigenic properties persisted postnatally in lCMN. Subpopulations of lCMN cells expressed stem cell/progenitor lineage markers such as Sox10, Nestin, Oct4, and ABCB5. In vitro, 1 in 250 cells from fresh lCMN formed colonies that could be passaged and harbored the same NRAS mutation as the original nevus. In vivo, lCMN specimens xenografted in immunocompromised mice expanded 4-fold. BrdU(+)-proliferating and label-retaining melanocytes were found within the outgrowth skin tissue of these xenografts, which displayed the same benign nested architecture as the original nevus. lCMN cell suspensions were not able to expand when xenografted alone in Rag 2-/- mice. Conversely, when mixed with keratinocytes, these cells reconstituted the architecture of the human nevus with its characteristic melanocyte layout, lentiginous hyperplasia, and nested architecture. Overall, our data demonstrate that, after birth, certain lCMN cell subtypes still display features such as clonogenic potential and expand into nevus-like structures when cooperating with adjacent keratinocytes.


Asunto(s)
Nevo Pigmentado/congénito , Nevo Pigmentado/patología , Neoplasias Cutáneas/congénito , Neoplasias Cutáneas/patología , Ensayo de Tumor de Célula Madre , Animales , Proliferación Celular , Transformación Celular Neoplásica , Preescolar , GTP Fosfohidrolasas/genética , Xenoinjertos , Proteínas de Homeodominio/genética , Humanos , Técnicas In Vitro , Lactante , Queratinocitos/patología , Melanocitos/patología , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Mutación/genética , Nevo Pigmentado/genética , Neoplasias Cutáneas/genética
7.
J Invest Dermatol ; 134(4): 1067-1074, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24129063

RESUMEN

Congenital melanocytic nevus (CMN) is a particular melanocytic in utero proliferation characterized by an increased risk of melanoma transformation during infancy or adulthood. NRAS and BRAF mutations have consistently been reported in CMN samples, but until recently results have been contradictory. We therefore studied a series of large and giant CMNs and compared them with small and medium CMNs using Sanger sequencing, pyrosequencing, high-resolution melting analysis, and mutation enrichment by an enhanced version of ice-COLD-PCR. Large-giant CMNs displayed NRAS mutations in 94.7% of cases (18/19). At that point, the role of additional mutations in CMN pathogenesis had to be investigated. We therefore performed exome sequencing on five specimens of large-giant nevi. The results showed that NRAS mutation was the sole recurrent somatic event found in such melanocytic proliferations. The genetic profile of small-medium CMNs was significantly different, with 70% of cases bearing NRAS mutations and 30% showing BRAF mutations. These findings strongly suggest that NRAS mutations are sufficient to drive melanocytic benign proliferations in utero.


Asunto(s)
GTP Fosfohidrolasas/genética , Genes ras , Proteínas de la Membrana/genética , Mutación , Nevo Pigmentado/congénito , Nevo Pigmentado/genética , Proliferación Celular , Niño , Preescolar , Análisis Mutacional de ADN , Exoma , Femenino , GTP Fosfohidrolasas/metabolismo , Genotipo , Humanos , Lactante , Masculino , Melanocitos/citología , Proteínas de la Membrana/metabolismo , Fenotipo , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Riesgo
8.
Genes Dev ; 25(8): 831-44, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21498572

RESUMEN

Neural stem cells (NSCs) are slowly dividing astrocytes that are intimately associated with capillary endothelial cells in the subventricular zone (SVZ) of the brain. Functionally, members of the vascular endothelial growth factor (VEGF) family can stimulate neurogenesis as well as angiogenesis, but it has been unclear whether they act directly via VEGF receptors (VEGFRs) expressed by neural cells, or indirectly via the release of growth factors from angiogenic capillaries. Here, we show that VEGFR-3, a receptor required for lymphangiogenesis, is expressed by NSCs and is directly required for neurogenesis. Vegfr3:YFP reporter mice show VEGFR-3 expression in multipotent NSCs, which are capable of self-renewal and are activated by the VEGFR-3 ligand VEGF-C in vitro. Overexpression of VEGF-C stimulates VEGFR-3-expressing NSCs and neurogenesis in the SVZ without affecting angiogenesis. Conversely, conditional deletion of Vegfr3 in neural cells, inducible deletion in subventricular astrocytes, and blocking of VEGFR-3 signaling with antibodies reduce SVZ neurogenesis. Therefore, VEGF-C/VEGFR-3 signaling acts directly on NSCs and regulates adult neurogenesis, opening potential approaches for treatment of neurodegenerative diseases.


Asunto(s)
Neurogénesis/fisiología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Linfangiogénesis/genética , Linfangiogénesis/fisiología , Ratones , Ratones Mutantes , Microscopía Electrónica de Transmisión , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/fisiología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética
9.
J Neuropathol Exp Neurol ; 69(8): 828-37, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20613635

RESUMEN

Inhaled nitric oxide (iNO) is one of the most promising therapies used in neonates, but there is little information available about its effect on the developing brain. We explored the effects of both iNO and endogenous NO on developing white matter in rodents. Rat or mouse pups and their mothers were placed in a chamber containing 5 to 20 ppm of NO for 7 days after birth. Neonatal exposure to iNO was associated with a transient increase in central nervous system myelination in rats and C57BL/6 mice without any deleterious effects at low doses (5 ppm) or behavioral consequences in adulthood. Exposure to iNO was associated with a proliferative effect on immature oligodendrocytes and a subsequent promaturational effect. The role of endogenous NO in myelination was investigated in animals treated with the nitric oxides synthase inhibitor N-nitro-L-arginine methyl ester (L-NAME) in the neonatal period; this led to protracted myelination defects and subsequent behavioral deficits in adulthood. These effects were reversed by rescuing L-NAME-treated animals with iNO. Thus, we demonstrate considerable effect of both exogenous and endogenous NO on myelination in rodents. These data point to potential new avenues for neuroprotection in human perinatal brain damage.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Depuradores de Radicales Libres/farmacología , Fibras Nerviosas Mielínicas/efectos de los fármacos , Óxido Nítrico/administración & dosificación , Óxido Nítrico/metabolismo , Administración por Inhalación , Factores de Edad , Animales , Animales Recién Nacidos , Antígenos/metabolismo , Síntomas Conductuales/inducido químicamente , Síntomas Conductuales/enzimología , Encéfalo/citología , Proliferación Celular/efectos de los fármacos , Sistema Nervioso Central/citología , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Conducta Exploratoria/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Etiquetado Corte-Fin in Situ/métodos , Antígeno Ki-67/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Básica de Mielina/metabolismo , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo , NG-Nitroarginina Metil Éster/farmacología , Fibras Nerviosas Mielínicas/metabolismo , Fibras Nerviosas Mielínicas/ultraestructura , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/ultraestructura , Pruebas Neuropsicológicas , Óxido Nítrico Sintasa de Tipo II/farmacología , Antígenos O/metabolismo , Oligodendroglía/efectos de los fármacos , Proteoglicanos/metabolismo , Ratas , Ratas Sprague-Dawley , Percepción Espacial/efectos de los fármacos , Conducta Espacial/efectos de los fármacos , Estadísticas no Paramétricas
10.
PLoS One ; 4(9): e7128, 2009 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-19771167

RESUMEN

OBJECTIVE: To investigate the effects of melatonin treatment in a rat model of white matter damage (WMD) in the developing brain. Additionally, we aim to delineate the cellular mechanisms of melatonin effect on the oligodendroglial cell lineage. METHODS: A unilateral ligation of the uterine artery in pregnant rat at the embryonic day 17 induces fetal hypoxia and subsequent growth restriction (GR) in neonatal pups. GR and control pups received a daily intra-peritoneal injection of melatonin from birth to post-natal day (P) 3. RESULTS: Melatonin administration was associated with a dramatic decrease in microglial activation and astroglial reaction compared to untreated GR pups. At P14, melatonin prevented white matter myelination defects with an increased number of mature oligodendrocytes (APC-immunoreactive) in treated GR pups. Conversely, melatonin was not found to be associated with an increased density of total oligodendrocytes (Olig2-immunoreactive), suggesting that melatonin is able to promote oligodendrocyte maturation but not proliferation. These effects appear to be melatonin-receptor dependent and were reproduced in vitro. INTERPRETATION: These data suggest that melatonin has a strong protective effect on developing damaged white matter through decreased microglial activation and oligodendroglial maturation leading to a normalization of the myelination process. Consequently, melatonin should be a considered as an effective neuroprotective candidate not only in perinatal brain damage but also in inflammatory and demyelinating diseases observed in adults.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Melatonina/fisiología , Fibras Nerviosas Mielínicas/metabolismo , Oligodendroglía/metabolismo , Animales , Animales Recién Nacidos , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Linaje de la Célula , Células Cultivadas , Inmunohistoquímica/métodos , Melatonina/metabolismo , Microscopía Fluorescente/métodos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
11.
Proc Natl Acad Sci U S A ; 105(43): 16779-84, 2008 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-18922769

RESUMEN

White-matter damage is a leading cause of neurological handicap. Although hypoxia-ischemia and excitotoxicity are major pathogenic factors, a role for genetic influences was suggested recently. Thus, protracted gestational hypoxia was associated with white-matter damage (WMD) in rat pups but not in mouse pups. Indeed, microglial activation and vessel-wall density on postnatal days (P)1 and P10 were found increased in both mouse and rat pups, but cell death, astrogliosis, and myelination were only significantly altered in hypoxic rat pups. We investigated whether this species-related difference was ascribable to effects of antenatal hypoxia on the expression of glutamate receptor subunits by using immunocytochemistry, PCR, and excitotoxic double hit insult. Quantitative PCR in hypoxic mouse pups on P1 showed 2- to 4-fold down-regulation of the AMPA-receptor subunits -1, 2, and -4; of the kainate-receptor subunit GluR7; and of the metabotropic receptor subunits mGluR1, -2, -3, -5, and -7. None of the glutamate-receptor subunits was down-regulated in the hypoxic rat pups. NR2B was the only NMDA-receptor subunit that was down-regulated in hypoxic mice but not in hypoxic rat on P1. Ifenprodil administration to induce functional inhibition of NMDA containing NR2B-subunit receptors prevented hypoxia-induced myelination delay in rat pups. Intracerebral injection of a glutamate agonist produced a larger decrease in ibotenate-induced excitotoxic lesions in hypoxic mouse pups than in normoxic mouse pups. Gestational hypoxia may regulate the expression of specific glutamate-receptor subunits in fetal mice but not in fetal rats. Therefore, genetic factors may influence the susceptibility of rodents to WMD.


Asunto(s)
Regulación de la Expresión Génica , Hipoxia/genética , Fibras Nerviosas Mielínicas/patología , Receptores de Glutamato/genética , Animales , Animales Recién Nacidos , Femenino , Predisposición Genética a la Enfermedad , Hipoxia/etiología , Ratones , Embarazo , Subunidades de Proteína/genética , Ratas , Receptores AMPA/genética , Receptores de Ácido Kaínico/genética , Receptores de Glutamato Metabotrópico/genética , Especificidad de la Especie
12.
Neurobiol Dis ; 26(1): 243-52, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17306552

RESUMEN

We studied hypoxic preconditioning (HxP) in the murine developing brain, focusing on the role for vascular endothelial growth factor (VEGF). Newborn mice were used as follows: (1) HxP (or normoxia) then intracerebral (i.c.) NMDA or AMPA-kainate agonist; (2) HxP then intraperitoneal (i.p.) anti-VEGFR2/Flk1 or anti-VEGFR1/Flt1 monoclonal blocking antibody (mAb) then i.c. NMDA/AMPA-kainate agonist; (3) i.p. VEGF then i.c. NMDA/AMPA-kainate agonist; and (4) in mutants lacking the hypoxia-responsive element (HRE) of the VEGF-A gene (VEGF( partial differential/ partial differential)) and their wild-type littermates (VEGF(+/+)), HxP followed by i.c. NMDA agonist. HxP reduced the size of NMDA-related cortical and AMPA-kainate-related cortical and white matter excitotoxic lesions. Anti-VEGFR2/Flk1 mAb prevented HxP-induced neuroprotection. VEGF produced dose-dependent reduction in cortical lesions. HxP did not prevent, but instead exacerbated, brain lesions in VEGF( partial differential/ partial differential) mutants. Thus, exogenous as well as endogenous VEGF reduces excitotoxic brain lesions in the developing mouse. The VEGF/VEGFR2/Flk1 pathway is involved in the neuroprotective response to HxP.


Asunto(s)
Animales Recién Nacidos/fisiología , Hipoxia Encefálica/fisiopatología , Factor A de Crecimiento Endotelial Vascular/fisiología , Envejecimiento/fisiología , Alanina/análogos & derivados , Alanina/farmacología , Animales , Anticuerpos Bloqueadores/farmacología , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Agonistas de Aminoácidos Excitadores , Hipoxia Encefálica/patología , Ácido Iboténico , Inyecciones Intraperitoneales , Ratones , Ratones Transgénicos , Mutación/fisiología , Regiones Promotoras Genéticas/genética , Pirimidinonas/farmacología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Uracilo/farmacología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Pérdida de Peso/efectos de los fármacos
13.
Pediatr Res ; 61(1): 54-60, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17211141

RESUMEN

In addition to glucose, monocarboxylates including lactate represent a major source of energy for the developing brain and appears to be crucial in the pathogenesis and recovery after brain damage. We hypothesized a role of monocarboxylates transport in the energy supply of neurons of the immature cerebral cortex. The effects of the blockade of monocarboxylates transport in vivo on the cortical development was investigated in neonatal mice using alpha-cyano-4-hydroxycinnamate (CIN) diluted either in DMSO (CD) or in ethanol (CE) administered intraperitoneally over postnatal day (P) P1 to P3. Injection of CIN induced a cytoarchitectonic disorganization in the parietal cortex likely due to a combination of slight disturbance of cortical neuronal migration and an increased neuronal cell death observed in CE (p < 0.05) but not in CD group. An increased number of activated GFAP-positive astroglia was observed in the neocortex in groups treated with CIN (CD and CE) on P10. These data: 1) Provide first evidence of deleterious effects observed in vivo after blockade of monocarboxylates transport in the developing brain; 2) emphasize the role of lactate during neuronal migration as a major source of energy; and 3) suggest the synergistic effect of ethanol-induced hypoglycemia in cortical brain damage induced by CIN.


Asunto(s)
Ácidos Carboxílicos/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/crecimiento & desarrollo , Ácidos Cumáricos/farmacología , Transportadores de Ácidos Monocarboxílicos/antagonistas & inhibidores , Animales , Animales Recién Nacidos , Corteza Cerebral/metabolismo , Ratones , Fenotipo
14.
Brain ; 129(Pt 12): 3209-23, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17028311

RESUMEN

Excitotoxic damage appears to be a critical factor in the formation of perinatal brain lesions associated with cerebral palsy (CP). When injected into newborn mice, the glutamatergic analogue, ibotenate, produces cortical lesions and white matter cysts that mimic human perinatal brain lesions. Neuropeptides are neuronal activity modulators and could therefore modulate glutamate-induced lesions. However, neuropeptides are rapidly degraded by peptidases. Racecadotril, which is rapidly metabolized to its active metabolite thiorphan, is a neutral endopeptidase (NEP) inhibitor used in clinical practice for diarrhoea with a remarkable safety profile. This study aimed to test the original hypothesis that thiorphan could be neuroprotective against ibotenate-induced lesions in newborn mice. Intraperitoneal administration of thiorphan reduced ibotenate-induced cortical lesions by up to 57% and cortical caspase-3 cleavage by up to 59%. This neuroprotective effect was long-lasting and was still observed when thiorphan was administered 12 h after the insult, showing a remarkable window for therapeutic intervention. Further supporting the neuroprotective effect of pharmacological blockade of NEP, mouse pups with a genetic deletion of NEP displayed a significantly reduced size of the ibotenate-induced cortical grey matter lesion when compared with wild-type animals. Thiorphan effects were mimicked by substance P (SP) and, in a less potent manner, by neurokinin A. Thiorphan effects were inhibited by blockers of NK1 and NK2 receptors. Real-time reverse transcription-polymerase chain reaction, autoradiography and immunohistochemistry confirmed the expression of NK1 and NK2 receptors in the neonatal murine neocortex. These data demonstrate that thiorphan prevents neonatal excitotoxic cortical damage, an effect largely mediated by SP. Thiorphan could represent a promising drug for the prevention of CP, which remains a challenging disease. In a broader context, these results also raise potential implications for the prevention of neurodegenerative diseases involving glutamate-mediated excitotoxic neuronal death.


Asunto(s)
Corteza Cerebral/efectos de los fármacos , Parálisis Cerebral/patología , Neprilisina/antagonistas & inhibidores , Fármacos Neuroprotectores/administración & dosificación , Inhibidores de Proteasas/administración & dosificación , Tiorfan/administración & dosificación , Animales , Animales Recién Nacidos , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/patología , Parálisis Cerebral/prevención & control , Agonistas de Aminoácidos Excitadores , Femenino , Ácido Iboténico , Inmunohistoquímica/métodos , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , Neuroquinina A/farmacología , Antagonistas del Receptor de Neuroquinina-1 , Neuronas/efectos de los fármacos , Neurotransmisores/farmacología , Receptores de Neuroquinina-2/antagonistas & inhibidores , Convulsiones/prevención & control , Sustancia P/farmacología
15.
Br J Pharmacol ; 148(4): 442-51, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16682966

RESUMEN

Brain lesions induced in newborn mice or rats by the glutamatergic agonists ibotenate (acting on NMDA and metabotropic receptors) or S-bromowillardiine (acting on AMPA-kainate receptors) mimic some aspects of white matter cysts and transcortical necrosis observed in human perinatal brain damage associated with cerebral palsy. Exogenous and endogenous cannabinoids have received increasing attention as potential neuroprotective agents in a number of neurodegenerative disorders of the adult. One recent study showed neuroprotection by the cannabinoid agonist WIN-55212 in a newborn rat model of acute severe asphyxia. The present study was designed to assess the neuroprotective effects of the endogenous cannabinoid anandamide using a well-defined rodent model of neonatal excitotoxic brain lesions. In this model, anandamide provided dose-dependent and long-lasting protection of developing white matter and cortical plate reducing the size of lesions induced by S-bromowillardiine. Anandamide had only marginal neuroprotective effect against ibotenate-induced cortical grey matter lesions. Anandamide-induced neuroprotection against AMPA-kainate receptor-mediated brain lesions were blocked by a CB1 antagonist but not by a CB2 antagonist. Furthermore, anandamide effects were mimicked by a CB1 agonist but not by a CB2 agonist. Real-time PCR confirmed the expression of CB1 receptors, but not CB2 receptors, in the untreated newborn neocortex. Finally, neuroprotective effects of anandamide in white matter involved increased survival of preoligodendrocytes and better preservation of myelination. The present study provides experimental support for the role of endocannabinoids as a candidate therapy for excitotoxic perinatal brain lesions.


Asunto(s)
Ácidos Araquidónicos/farmacología , Encéfalo/efectos de los fármacos , Moduladores de Receptores de Cannabinoides/farmacología , Endocannabinoides , Fármacos Neuroprotectores/farmacología , Receptores AMPA/fisiología , Receptores de Ácido Kaínico/fisiología , Animales , Animales Recién Nacidos , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Ácido Iboténico/farmacología , Masculino , Ratones , Alcamidas Poliinsaturadas , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB2/genética
16.
Neurobiol Dis ; 18(1): 193-205, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15649710

RESUMEN

Intraneocortical injection of ibotenate, a glutamate analog, in newborn mice produces damage mimicking lesions observed in human infants with cerebral palsy. Previous research using this model has demonstrated that pretreatment with IL-9, a Th2 cytokine, significantly exacerbated excitotoxic brain lesions. The goal of this study is to identify the underlying pathophysiological mechanism of lesion formation. Pretreatment with TGF-beta1 produced the same effects as IL-9 on ibotenate-induced lesions. IL-9 effects were abolished when a specific TGF-beta1 neutralizing antibody is administered at the same time. Real-time PCR, Western blot, and immunohistochemistry showed that pretreatment with IL-9 increased TGF-beta1 neocortical expression. In vitro studies using real-time PCR and immunocytochemistry demonstrated that neurons were a major contributor in IL-9-induced increase of TGF-beta1. In c-Kit mast cell-deficient mice, TGF-beta1 failed to exacerbate excitotoxic brain lesions, suggesting a key role of mast cells in TGF-beta1 effects. A specific inhibitor of mast cell degranulation and histamine receptor blockers abrogated TGF-beta1 effects on excitotoxic lesions, providing further evidence of mast cell involvement and the role of mast cell-derived histamine. Finally, in vitro studies using a mast cell line showed that TGF-beta1 increased histamine in the supernatant. In aggregate, these data support the notion that neuronal TGF-beta1 plays a key role in the IL-9/mast cell interaction, which leads to an exacerbation of neonatal excitotoxic damage through an increased extracellular histamine concentration. The identification of this pathway, if confirmed in human neonates, might have important implications for understanding and preventing cerebral palsy.


Asunto(s)
Parálisis Cerebral/inmunología , Interleucina-9/toxicidad , Mastocitos/inmunología , Degeneración Nerviosa/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Animales Recién Nacidos , Anticuerpos/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/metabolismo , Línea Celular , Parálisis Cerebral/metabolismo , Parálisis Cerebral/fisiopatología , Modelos Animales de Enfermedad , Femenino , Histamina/metabolismo , Antagonistas de los Receptores Histamínicos/farmacología , Interleucina-9/inmunología , Masculino , Mastocitos/efectos de los fármacos , Mastocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/metabolismo , Neuronas/inmunología , Neuronas/metabolismo , Neurotoxinas/inmunología , Neurotoxinas/metabolismo , Receptores Histamínicos/efectos de los fármacos , Receptores Histamínicos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta1
17.
Brain Pathol ; 14(1): 1-10, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14997932

RESUMEN

In the premature infant, periventricular leukomalacia, usually related to hypoxicischemic white matter damage, is the main cause of neurological impairment. We hypothesized that protracted prenatal hypoxia might induce white matter damage during the perinatal period. Pregnant Sprague-Dawley rats were placed in a chamber supplied with hypoxic gas (10% O2-90% N2) from embryonic day 5 (E5) to E20. Neonatal rat brains were investigated by histology, immunocytochemistry, western blotting, in situ hybridization, DNA fragmentation analysis, and in vivo magnetic resonance imaging (MRI). Body weight of pups subjected to prenatal hypoxia was 10 to 30% lower from P0 to P14 than in controls. Specific white matter cysts were detected between P0 and P7 in pups subjected to prenatal hypoxia, in addition to abnormal extra-cellular matrix, increased lipid peroxidation, white matter cell death detected by TUNEL, and increased activated macrophage counts in white matter. Subsequently, gliotic scars and delayed myelination primarily involving immature oligodendrocytes were seen. In vivo MRI with T1, T2, and diffusion sequences disclosed similar findings immediately after birth, showing strong correlations with histological abnormalities. We speculate that protracted prenatal hypoxia in rat induces white matter damage occurring through local inflammatory response and oxidative stress linked to re-oxygenation during the perinatal period.


Asunto(s)
Encéfalo/patología , Hipoxia/patología , Leucomalacia Periventricular/etiología , Efectos Tardíos de la Exposición Prenatal , Animales , Animales Recién Nacidos , Western Blotting , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunohistoquímica , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Recién Nacido , Leucomalacia Periventricular/patología , Imagen por Resonancia Magnética , Embarazo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...