Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Int J Mol Sci ; 22(17)2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34502373

RESUMEN

Rett syndrome is a neurodevelopmental disorder caused by mutations of the methyl-CpG binding protein 2 gene. Abnormal physiological functions of glial cells contribute to pathogenesis of Rett syndrome. Semaphorin 4D (SEMA4D) regulates processes central to neuroinflammation and neurodegeneration including cytoskeletal structures required for process extension, communication, and migration of glial cells. Blocking SEMA4D-induced gliosis may preserve normal glial and neuronal function and rescue neurological dysfunction in Rett syndrome. We evaluated the pre-clinical therapeutic efficacy of an anti-SEMA4D monoclonal antibody in the Rett syndrome Mecp2T158A transgenic mouse model and investigated the contribution of glial cells as a proposed mechanism of action in treated mice and in primary glial cultures isolated from Mecp2T158A/y mutant mice. SEMA4D is upregulated in neurons while glial fibrillary acidic protein and ionized calcium binding adaptor molecule 1-positive cells are upregulated in Mecp2T158A/y mice. Anti-SEMA4D treatment ameliorates Rett syndrome-specific symptoms and improves behavioural functions in both pre-symptomatic and symptomatic cohorts of hemizygous Mecp2T158A/y male mice. Anti-SEMA4D also reduces astrocyte and microglia activation in vivo. In vitro experiments demonstrate an abnormal cytoskeletal structure in mutant astrocytes in the presence of SEMA4D, while anti-SEMA4D antibody treatment blocks SEMA4D-Plexin B1 signaling and mitigates these abnormalities. These results suggest that anti-SEMA4D immunotherapy may be an effective treatment option to alleviate symptoms and improve cognitive and motor function in Rett syndrome.


Asunto(s)
Síndrome de Rett/metabolismo , Síndrome de Rett/fisiopatología , Semaforinas/metabolismo , Animales , Antígenos CD/inmunología , Antígenos CD/metabolismo , Cognición/fisiología , Modelos Animales de Enfermedad , Inmunoterapia , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Destreza Motora/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Receptores de Superficie Celular/metabolismo , Respiración/inmunología , Síndrome de Rett/genética , Semaforinas/genética , Semaforinas/inmunología , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
2.
FASEB J ; 31(6): 2327-2339, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28213359

RESUMEN

The eyes absent (EYA) family proteins are conserved transcriptional coactivators with intrinsic protein phosphatase activity. They play an essential role in the development of various organs in metazoans. These functions are associated with a unique combination of phosphatase and transactivation activities. However, it remains poorly understood how these activities and the consequent biologic functions of EYA are regulated. Here, we demonstrate that 2 conserved arginine residues, R304 and R306, of EYA1 are essential for its in vitro phosphatase activity and in vivo function during Drosophila eye development. EYA1 physically interacts with protein arginine methyltransferase 1, which methylates EYA1 at these residues both in vitro and in cultured mammalian and insect cells. Moreover, we show that wild-type, but not methylation-defective, EYA1 associates with γ-H2A.X in response to ionizing radiation. Taken together, our results identify the conserved arginine residues of EYA1 that play an important role for its activity, thus implicating arginine methylation as a novel regulatory mechanism of EYA function.-Li, X., Eberhardt, A., Hansen, J. N., Bohmann, D., Li, H., Schor, N. F. Methylation of the phosphatase-transcription activator EYA1 by protein arginine methyltransferase 1: mechanistic, functional, and structural studies.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Drosophila melanogaster , Regulación Enzimológica de la Expresión Génica , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Metilación , Mutación , Proteínas Nucleares/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Tirosina Fosfatasas/genética , Proteína-Arginina N-Metiltransferasas/genética
3.
Sci Transl Med ; 12(536)2020 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-32213628

RESUMEN

Despite high metabolic activity, the retina and optic nerve head lack traditional lymphatic drainage. We here identified an ocular glymphatic clearance route for fluid and wastes via the proximal optic nerve in rodents. ß-amyloid (Aß) was cleared from the retina and vitreous via a pathway dependent on glial water channel aquaporin-4 (AQP4) and driven by the ocular-cranial pressure difference. After traversing the lamina barrier, intra-axonal Aß was cleared via the perivenous space and subsequently drained to lymphatic vessels. Light-induced pupil constriction enhanced efflux, whereas atropine or raising intracranial pressure blocked efflux. In two distinct murine models of glaucoma, Aß leaked from the eye via defects in the lamina barrier instead of directional axonal efflux. The results suggest that, in rodents, the removal of fluid and metabolites from the intraocular space occurs through a glymphatic pathway that might be impaired in glaucoma.


Asunto(s)
Sistema Glinfático , Péptidos beta-Amiloides/metabolismo , Animales , Acuaporina 4/metabolismo , Sistema Glinfático/metabolismo , Presión Intracraneal , Ratones , Nervio Óptico , Retina , Cuerpo Vítreo
4.
Sci Rep ; 8(1): 2246, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29396480

RESUMEN

Prolonged intake of excessive amounts of ethanol is known to have adverse effects on the central nervous system (CNS). Here we investigated the effects of acute and chronic ethanol exposure and withdrawal from chronic ethanol exposure on glymphatic function, which is a brain-wide metabolite clearance system connected to the peripheral lymphatic system. Acute and chronic exposure to 1.5 g/kg (binge level) ethanol dramatically suppressed glymphatic function in awake mice. Chronic exposure to 1.5 g/kg ethanol increased GFAP expression and induced mislocation of the astrocyte-specific water channel aquaporin 4 (AQP4), but decreased the levels of several cytokines. Surprisingly, glymphatic function increased in mice treated with 0.5 g/kg (low dose) ethanol following acute exposure, as well as after one month of chronic exposure. Low doses of chronic ethanol intake were associated with a significant decrease in GFAP expression, with little change in the cytokine profile compared with the saline group. These observations suggest that ethanol has a J-shaped effect on the glymphatic system whereby low doses of ethanol increase glymphatic function. Conversely, chronic 1.5 g/kg ethanol intake induced reactive gliosis and perturbed glymphatic function, which possibly may contribute to the higher risk of dementia observed in heavy drinkers.


Asunto(s)
Consumo de Bebidas Alcohólicas/efectos adversos , Alcoholismo/patología , Etanol/administración & dosificación , Etanol/farmacología , Sistema Glinfático/efectos de los fármacos , Animales , Acuaporina 4/efectos de los fármacos , Citocinas/sangre , Demencia/inducido químicamente , Gliosis/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Sueño/fisiología
5.
J Cancer Res Ther (Manch) ; 4(2): 11-18, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28713571

RESUMEN

Neuroblastoma, the most frequently occurring extracranial solid tumor of childhood, arises from neural crest-derived cells that are arrested at an early stage of differentiation in the developing sympathetic nervous system. There is an urgent need to identify clinically relevant biomarkers for better prognosis and treatment of this aggressive malignancy. Eyes Absent 1 (EYA1) is an essential transcriptional coactivator for neuronal developmental programs during organogenesis. Whether or not EYA1 is implicated in neuroblastoma and subcellular localization of EYA1 is relevant to clinical behaviour of neuroblastoma is not known. We studied EYA1 expression and subcellular localization by immunohistochemistry in tissue microarrays containing tumor specimens from 98 patients, 66 of which were characterized by known clinical prognostic markers of neuroblastoma. Immunostaining results were evaluated and statistically correlated with the degree of histologic differentiation and with neuroblastoma risk stratification group characteristics, including stage of disease, patient age, tumor histology and mitosis-karyorrhexis index (MKI), respectively. We found that EYA1 levels were significantly higher in neuroblastomas than in ganglioneuromas and ganglioneuroblastomas. EYA1 was more highly expressed in stage 1,2,3 or 4S tumors as compared to stage 4 tumors (P<0.01). Tumors with high levels of nuclear EYA1 were more frequently associated with high nuclear MYCN levels. These results suggest that modulation of expression and intracellular localization of EYA1 in neural crest cells may provide a novel direction for therapeutic strategies.

6.
Oncotarget ; 7(39): 63629-63639, 2016 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-27571165

RESUMEN

Amplification or overexpression of MYCN is associated with poor prognosis of human neuroblastoma. We have recently defined a MYCN-dependent transcriptional signature, including protein arginine methyltransferase 1 (PRMT1), which identifies a subgroup of patients with high-risk disease. Here we provide several lines of evidence demonstrating PRMT1 as a novel regulator of MYCN and implicating PRMT1 as a potential therapeutic target in neuroblastoma pathogenesis. First, we observed a strong correlation between MYCN and PRMT1 protein levels in primary neuroblastoma tumors. Second, MYCN physically associates with PRMT1 by direct protein-protein interaction. Third, depletion of PRMT1 through siRNA knockdown reduced neuroblastoma cell viability and MYCN expression. Fourth, we showed that PRMT1 regulates MYCN stability and identified MYCN as a novel substrate of PRMT1. Finally, we demonstrated that mutation of putatively methylated arginine R65 to alanine decreased MYCN stability by altering phosphorylation at residues serine 62 and threonine 58. These results provide mechanistic insights into the modulation of MYCN oncoprotein by PRMT1, and suggest that targeting PRMT1 may have a therapeutic impact on MYCN-driven oncogenesis.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteína Proto-Oncogénica N-Myc/metabolismo , Neuroblastoma/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Arginina/química , Perfilación de la Expresión Génica , Humanos , Mutación , Fosforilación , Pronóstico , Modelos de Riesgos Proporcionales , Procesamiento Proteico-Postraduccional , ARN Interferente Pequeño/metabolismo , Treonina/química
7.
PLoS One ; 10(7): e0132375, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26177509

RESUMEN

Cytogenetically normal acute myeloid leukemia (CN-AML) patients harboring RUNX1 mutations have a dismal prognosis with anthracycline/cytarabine-based chemotherapy. We aimed to develop an in vivo model of RUNX1-mutated, CN-AML in which the nature of residual disease in this molecular disease subset could be explored. We utilized a well-characterized patient-derived, RUNX1-mutated CN-AML line (CG-SH). Tail vein injection of CG-SH into NOD scid gamma mice led to leukemic engraftment in the bone marrow, spleen, and peripheral blood within 6 weeks. Treatment of leukemic mice with anthracycline/cytarabine-based chemotherapy resulted in clearance of disease from the spleen and peripheral blood, but persistence of disease in the bone marrow as assessed by flow cytometry and secondary transplantation. Whole exome sequencing of CG-SH revealed mutations in ASXL1, CEBPA, GATA2, and SETBP1, not previously reported. We conclude that CG-SH xenografts are a robust, reproducible in vivo model of CN-AML in which to explore mechanisms of chemotherapy resistance and novel therapeutic approaches.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Análisis Citogenético , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Mutación/genética , Neoplasia Residual/genética , Neoplasia Residual/patología , Animales , Antraciclinas/farmacología , Antraciclinas/uso terapéutico , Médula Ósea/patología , Línea Celular Tumoral , Citarabina/farmacología , Citarabina/uso terapéutico , Análisis Mutacional de ADN , Exoma/genética , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Ratones Endogámicos NOD , Ratones SCID , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA