Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Hum Mol Genet ; 28(15): 2589-2599, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31035284

RESUMEN

The SOXC transcription factors Sox4, Sox11 and Sox12, are critical neurodevelopmental regulators that are thought to function in a highly redundant fashion. Surprisingly, heterozygous missense mutations or deletions of SOX11 were recently detected in patients with Coffin-Siris syndrome-like syndrome (CSSLS), a neurodevelopmental disorder associated with intellectual disability, demonstrating that in humans SOX11 haploinsufficiency cannot be compensated and raising the question of the function of SOX11 in human neurodevelopment. Here, we describe the generation of SOX11+/- heterozygous human embryonic stem cell (hESC) lines by CRISPR/Cas9 genome engineering. SOX11 haploinsufficiency impaired the generation of neurons and resulted in a proliferation/differentiation imbalance of neural precursor cells and enhanced neuronal cell death. Using the SOX11+/- hESC model we provide for the first time experimental evidence that SOX11 haploinsufficiency is sufficient to impair key processes of human neurodevelopment, giving a first insight into the pathophysiology of CSSLS and SOX11 function in human neurodevelopment.


Asunto(s)
Línea Celular , Dosificación de Gen , Células Madre Embrionarias Humanas/fisiología , Modelos Biológicos , Trastornos del Neurodesarrollo/metabolismo , Factores de Transcripción SOXC/genética , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Sistemas CRISPR-Cas , Diferenciación Celular , Proliferación Celular , Cara/anomalías , Edición Génica , Regulación de la Expresión Génica , Deformidades Congénitas de la Mano/genética , Deformidades Congénitas de la Mano/metabolismo , Haploinsuficiencia , Células Madre Embrionarias Humanas/metabolismo , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Micrognatismo/genética , Micrognatismo/metabolismo , Cuello/anomalías , Células-Madre Neurales , Trastornos del Neurodesarrollo/genética
2.
Brain Behav Immun ; 49: 156-70, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26074461

RESUMEN

Chronic peripheral inflammation mediated by cytokines such as TNFα, IL-1ß, and IL-6 is associated with psychiatric disorders like depression and anxiety. However, it remains elusive which distinct type of peripheral inflammation triggers neuroinflammation and affects hippocampal plasticity resulting in depressive-like behavior. We hypothesized that chronic peripheral inflammation in the human TNF-α transgenic (TNFtg) mouse model of rheumatoid arthritis spreads into the central nervous system and induces depressive state manifested in specific behavioral pattern and impaired adult hippocampal neurogenesis. TNFtg mice showed severe erosive arthritis with increased IL-1ß and IL-6 expression in tarsal joints with highly elevated human TNF-α levels in the serum. Intriguingly, IL-1ß and IL-6 mRNA levels were not altered in the hippocampus of TNFtg mice. In contrast to the pronounced monocytosis in joints and spleen of TNFtg mice, signs of hippocampal microgliosis or astrocytosis were lacking. Furthermore, locomotion was impaired, but there was no locomotion-independent depressive behavior in TNFtg mice. Proliferation and maturation of hippocampal neural precursor cells as well as survival of newly generated neurons were preserved in the dentate gyrus of TNFtg mice despite reduced motor activity and peripheral inflammatory signature. We conclude that peripheral inflammation in TNFtg mice is mediated by chronic activation of the innate immune system. However, severe peripheral inflammation, though impairing locomotor activity, does not elicit depressive-like behavior. These structural and functional findings indicate the maintenance of hippocampal immunity, cellular plasticity, and behavior despite peripheral innate inflammation.


Asunto(s)
Trastorno Depresivo/inmunología , Encefalitis/inmunología , Hipocampo/inmunología , Inmunidad Innata , Inflamación/inmunología , Animales , Artritis/genética , Artritis/inmunología , Trastorno Depresivo/etiología , Encefalitis/etiología , Femenino , Humanos , Inflamación/complicaciones , Inflamación/genética , Mediadores de Inflamación/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/inmunología , Células-Madre Neurales/inmunología , Neurogénesis/inmunología , Neuronas/inmunología , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/genética
3.
Front Neurosci ; 15: 680572, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34326717

RESUMEN

Pathogenic variants in SPG11 are the most frequent cause of autosomal recessive complicated hereditary spastic paraplegia (HSP). In addition to spastic paraplegia caused by corticospinal degeneration, most patients are significantly affected by progressive weakness and muscle wasting due to alpha motor neuron (MN) degeneration. Mitochondria play a crucial role in neuronal health, and mitochondrial deficits were reported in other types of HSPs. To investigate whether mitochondrial pathology is present in SPG11, we differentiated MNs from induced pluripotent stem cells derived from SPG11 patients and controls. MN derived from human embryonic stem cells and an isogenic SPG11 knockout line were also included in the study. Morphological analysis of mitochondria in the MN soma versus neurites revealed specific alterations of mitochondrial morphology within SPG11 neurites, but not within the soma. In addition, impaired mitochondrial membrane potential was indicative of mitochondrial dysfunction. Moreover, we reveal neuritic aggregates further supporting neurite pathology in SPG11. Correspondingly, using a microfluidic-based MN culture system, we demonstrate that axonal mitochondrial transport was significantly impaired in SPG11. Overall, our data demonstrate that alterations in morphology, function, and transport of mitochondria are an important feature of axonal dysfunction in SPG11 MNs.

4.
Stem Cell Reports ; 10(2): 347-355, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29337116

RESUMEN

Swiprosin-1/Efhd2 (Efhd2) is highly expressed in the CNS during development and in the adult. EFHD2 is regulated by Ca2+ binding, stabilizes F-actin, and promotes neurite extension. Previous studies indicated a dysregulation of EFHD2 in human Alzheimer's disease brains. We hypothesized a detrimental effect of genetic ablation of Efhd2 on hippocampal integrity and specifically investigated adult hippocampal neurogenesis. Efhd2 was expressed throughout adult neuronal development and in mature neurons. We observed a severe reduction of the survival of adult newborn neurons in Efhd2 knockouts, starting at the early neuroblast stage. Spine formation and dendrite growth of newborn neurons were compromised in full Efhd2 knockouts, but not upon cell-autonomous Efhd2 deletion. Together with our finding of severe hippocampal tauopathy in Efhd2 knockout mice, these data connect Efhd2 to impaired synaptic plasticity as present in Alzheimer's disease and identify a role of Efhd2 in neuronal survival and synaptic integration in the adult hippocampus.


Asunto(s)
Enfermedad de Alzheimer/genética , Proteínas de Unión al Calcio/genética , Hipocampo/crecimiento & desarrollo , Neurogénesis/genética , Columna Vertebral/crecimiento & desarrollo , Actinas/genética , Enfermedad de Alzheimer/patología , Animales , Calcio/metabolismo , Sistema Nervioso Central/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica/genética , Hipocampo/metabolismo , Humanos , Ratones , Ratones Noqueados , Neuritas/metabolismo , Plasticidad Neuronal/genética , Neuronas/citología , Neuronas/metabolismo , Columna Vertebral/metabolismo
5.
Brain Struct Funct ; 223(3): 1357-1368, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29124353

RESUMEN

In the adult mammalian hippocampus, new neurons are constantly added to the dentate gyrus. Adult neurogenesis is impaired in several neurodegenerative mouse models including α-synuclein (a-syn) transgenic mice. Among different a-syn species, a-syn oligomers were reported to be the most toxic species for neurons. Here, we studied the impact of wild-type vs. oligomer-prone a-syn on neurogenesis. We compared the wild-type a-syn transgenic mouse model (Thy1-WTS) to its equivalent transgenic for oligomer-prone E57K-mutant a-syn (Thy1-E57K). Transgenic a-syn was highly expressed within the hippocampus of both models, but was not present within adult neural stem cells and neuroblasts. Proliferation and survival of newly generated neurons were unchanged in both transgenic models. Thy1-WTS showed a minor integration deficit regarding mushroom spine density of newborn neurons, whereas Thy1-E57K exhibited a severe reduction of all spines. We conclude that cell-extrinsic a-syn impairs mushroom spine formation of adult newborn neurons and that oligomer-prone a-syn exacerbates this integration deficit. Moreover, our data suggest that a-syn reduces the survival of newborn neurons by a cell-intrinsic mechanism during the early neuroblast development. The finding of increased spine pathology in Thy1-E57K is a new pathogenic function of oligomeric a-syn and precedes overt neurodegeneration. Thus, it may constitute a readout for therapeutic approaches.


Asunto(s)
Hipocampo/patología , Mutación/genética , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Neurogénesis/genética , alfa-Sinucleína/genética , Animales , Bromodesoxiuridina/metabolismo , Caspasa 3/metabolismo , Proliferación Celular/genética , Modelos Animales de Enfermedad , Proteínas de Dominio Doblecortina , Regulación de la Expresión Génica/genética , Ácido Glutámico/genética , Lisina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/patología , Neuronas/ultraestructura , Neuropéptidos/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Factores de Transcripción SOXB1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA