Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Acta Neuropathol ; 147(1): 107, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918213

RESUMEN

Alzheimer's disease (AD) is the most common cause of dementia, and disease mechanisms are still not fully understood. Here, we explored pathological changes in human induced pluripotent stem cell (iPSC)-derived neurons carrying the familial AD APPV717I mutation after cell injection into the mouse forebrain. APPV717I mutant iPSCs and isogenic controls were differentiated into neurons revealing enhanced Aß42 production, elevated phospho-tau, and impaired neurite outgrowth in APPV717I neurons. Two months after transplantation, APPV717I and control neural cells showed robust engraftment but at 12 months post-injection, APPV717I grafts were smaller and demonstrated impaired neurite outgrowth compared to controls, while plaque and tangle pathology were not seen. Single-nucleus RNA-sequencing of micro-dissected grafts, performed 2 months after cell injection, identified significantly altered transcriptome signatures in APPV717I iPSC-derived neurons pointing towards dysregulated synaptic function and axon guidance. Interestingly, APPV717I neurons showed an increased expression of genes, many of which are also upregulated in postmortem neurons of AD patients including the transmembrane protein LINGO2. Downregulation of LINGO2 in cultured APPV717I neurons rescued neurite outgrowth deficits and reversed key AD-associated transcriptional changes related but not limited to synaptic function, apoptosis and cellular senescence. These results provide important insights into transcriptional dysregulation in xenografted APPV717I neurons linked to synaptic function, and they indicate that LINGO2 may represent a potential therapeutic target in AD.


Asunto(s)
Enfermedad de Alzheimer , Precursor de Proteína beta-Amiloide , Células Madre Pluripotentes Inducidas , Neuronas , Transcriptoma , Humanos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/metabolismo , Neuronas/metabolismo , Neuronas/patología , Animales , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Mutación , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Sinapsis/patología , Sinapsis/metabolismo , Péptidos beta-Amiloides/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología
2.
Cell Stem Cell ; 31(5): 676-693.e10, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38626772

RESUMEN

Frontotemporal dementia (FTD) is an incurable group of early-onset dementias that can be caused by the deposition of hyperphosphorylated tau in patient brains. However, the mechanisms leading to neurodegeneration remain largely unknown. Here, we combined single-cell analyses of FTD patient brains with a stem cell culture and transplantation model of FTD. We identified disease phenotypes in FTD neurons carrying the MAPT-N279K mutation, which were related to oxidative stress, oxidative phosphorylation, and neuroinflammation with an upregulation of the inflammation-associated protein osteopontin (OPN). Human FTD neurons survived less and elicited an increased microglial response after transplantation into the mouse forebrain, which we further characterized by single nucleus RNA sequencing of microdissected grafts. Notably, downregulation of OPN in engrafted FTD neurons resulted in improved engraftment and reduced microglial infiltration, indicating an immune-modulatory role of OPN in patient neurons, which may represent a potential therapeutic target in FTD.


Asunto(s)
Demencia Frontotemporal , Neuronas , Osteopontina , Proteínas tau , Osteopontina/metabolismo , Osteopontina/genética , Demencia Frontotemporal/genética , Demencia Frontotemporal/patología , Demencia Frontotemporal/metabolismo , Humanos , Neuronas/metabolismo , Neuronas/patología , Animales , Proteínas tau/metabolismo , Ratones , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/patología , Microglía/metabolismo , Microglía/patología , Mutación/genética
3.
Front Aging Neurosci ; 16: 1354164, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38450383

RESUMEN

Alzheimer's disease (AD) is the most frequent form of dementia. It is characterized by pronounced neuronal degeneration with formation of neurofibrillary tangles and deposition of amyloid ß throughout the central nervous system. Animal models have provided important insights into the pathogenesis of AD and they have shown that different brain cell types including neurons, astrocytes and microglia have important functions in the pathogenesis of AD. However, there are difficulties in translating promising therapeutic observations in mice into clinical application in patients. Alternative models using human cells such as human induced pluripotent stem cells (iPSCs) may provide significant advantages, since they have successfully been used to model disease mechanisms in neurons and in glial cells in neurodegenerative diseases in vitro and in vivo. In this review, we summarize recent studies that describe the transplantation of human iPSC-derived neurons, astrocytes and microglial cells into the forebrain of mice to generate chimeric transplantation models of AD. We also discuss opportunities, challenges and limitations in using differentiated human iPSCs for in vivo disease modeling and their application for biomedical research.

4.
Brain Pathol ; : e13231, 2024 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-38246596

RESUMEN

Neurodegenerative diseases are increasing in prevalence and comprise a large socioeconomic burden on patients and their caretakers. The need for effective therapies and avenues for disease prevention and monitoring is of paramount importance. Fluid biomarkers for neurodegenerative diseases have gained a variety of uses, including informing participant selection for clinical trials, lending confidence to clinical diagnosis and disease staging, determining prognosis, and monitoring therapeutic response. Their role is expected to grow as disease-modifying therapies start to be available to a broader range of patients and as prevention strategies become established. Many of the underlying molecular mechanisms of currently used biomarkers are incompletely understood. Animal models and in vitro systems using cell lines have been extensively employed but face important translatability limitations. Induced pluripotent stem cell (iPSC) technology, where a theoretically unlimited range of cell types can be reprogrammed from peripheral cells sampled from patients or healthy individuals, has gained prominence over the last decade. It is a promising avenue to study physiological and pathological biomarker function and response to experimental therapeutics. Such systems are amenable to high-throughput drug screening or multiomics readouts such as transcriptomics, lipidomics, and proteomics for biomarker discovery, investigation, and validation. The present review describes the current state of biomarkers in the clinical context of neurodegenerative diseases, with a focus on Alzheimer's disease and frontotemporal dementia. We include a discussion of how iPSC models have been used to investigate and test biomarkers such as amyloid-ß, phosphorylated tau, neurofilament light chain or complement proteins, and even nominate novel biomarkers. We discuss the limitations of current iPSC methods, mentioning alternatives such as coculture systems and three-dimensional organoids which address some of these concerns. Finally, we propose exciting prospects for stem cell transplantation paradigms using animal models as a preclinical tool to study biomarkers in the in vivo context.

6.
Acta Neuropathol Commun ; 9(1): 160, 2021 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-34587990

RESUMEN

Infratentorial oligodendrogliomas, a rare pathological entity, are generally considered metastatic lesions from supratentorial primary tumors. Here, we report the case of a 23-year-old man presenting with a histopathologically confirmed right precentral gyrus grade 2 oligodendroglioma and a concurrent pontine grade 3 oligodendroglioma. The pontine lesion was biopsied approximately a year after the biopsy of the precentral lesion due to disease progression despite 4 cycles of procarbazine-CCNU-vincristine (PCV) chemotherapy and stable supratentorial disease. Histology and genetic analysis of the pontine biopsy were consistent with grade 3 oligodendroglioma, and comparison of the two lesions demonstrated common 1p/19q co-deletions and TERT promoter mutations but distinct IDH1 mutations, with a non-canonical IDH1 R132G mutation identified in the infratentorial lesion and a R132H mutation identified in the cortical lesion. Initiation of Temozolomide led to complete response of the supratentorial lesion and durable disease control, while Temozolomide with subsequent radiation therapy of 54 Gy in 30 fractions resulted in partial response of the pontine lesion. This case report supports possible distinct molecular pathogenesis in supratentorial and infratentorial oligodendrogliomas and raises questions about the role of different IDH1 mutant isoforms in explaining treatment resistance to different chemotherapy regimens. Importantly, this case suggests that biopsies of all radiographic lesions, when feasible and safe, should be considered in order to adequately guide management in multicentric oligodendrogliomas.


Asunto(s)
Neoplasias Encefálicas/genética , Isocitrato Deshidrogenasa/genética , Neoplasias Primarias Múltiples/genética , Oligodendroglioma/genética , Neoplasias Encefálicas/patología , Humanos , Masculino , Mutación , Neoplasias Primarias Múltiples/patología , Oligodendroglioma/patología , Adulto Joven
7.
Elife ; 102021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33904394

RESUMEN

Cortical interneurons establish inhibitory microcircuits throughout the neocortex and their dysfunction has been implicated in epilepsy and neuropsychiatric diseases. Developmentally, interneurons migrate from a distal progenitor domain in order to populate the neocortex - a process that occurs at a slower rate in humans than in mice. In this study, we sought to identify factors that regulate the rate of interneuron maturation across the two species. Using embryonic mouse development as a model system, we found that the process of initiating interneuron migration is regulated by blood vessels of the medial ganglionic eminence (MGE), an interneuron progenitor domain. We identified two endothelial cell-derived paracrine factors, SPARC and SerpinE1, that enhance interneuron migration in mouse MGE explants and organotypic cultures. Moreover, pre-treatment of human stem cell-derived interneurons (hSC-interneurons) with SPARC and SerpinE1 prior to transplantation into neonatal mouse cortex enhanced their migration and morphological elaboration in the host cortex. Further, SPARC and SerpinE1-treated hSC-interneurons also exhibited more mature electrophysiological characteristics compared to controls. Overall, our studies suggest a critical role for CNS vasculature in regulating interneuron developmental maturation in both mice and humans.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Corteza Cerebral/metabolismo , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Interneuronas/efectos de los fármacos , Eminencia Media/irrigación sanguínea , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Osteonectina/farmacología , Inhibidor 1 de Activador Plasminogénico/farmacología , Potenciales de Acción , Animales , Corteza Cerebral/embriología , Corteza Cerebral/cirugía , Células Endoteliales/metabolismo , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Interneuronas/metabolismo , Interneuronas/trasplante , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Eminencia Media/embriología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Neovascularización Fisiológica , Células-Madre Neurales/metabolismo , Células-Madre Neurales/trasplante , Osteonectina/metabolismo , Comunicación Paracrina , Inhibidor 1 de Activador Plasminogénico/metabolismo , Transducción de Señal
9.
J Neurosurg ; 128(2): 524-529, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28298028

RESUMEN

Tufted angioma (TA) is a rare, slow-growing, vascular lesion that commonly presents as a solitary macule, papule, or nodule arising in the soft tissues of the torso, extremities, and head and neck in children and young adults. Adult-onset cases have been infrequently reported. While typically benign, TAs may be locally aggressive. Complete physical examination and hematological workup are recommended in patients with TA to exclude the presence of Kasabach-Merritt phenomenon (KMP). The authors describe the case of a 69-year-old man with a contrast-enhancing frontal lobe lesion, with surrounding vasogenic edema, which was treated by gross-total resection. Characteristic histological features of a TA were demonstrated, with multiple cannonball-like tufts of densely packed capillaries emanating from intraparenchymal vessels in cerebral cortex and adjacent white matter. Tumor recurrence was detected after 4 months and treated with adjuvant Gamma Knife radiosurgery. To the extent of the authors' knowledge, this case illustrates the first report of TA presenting in an adult as an intracranial intraaxial tumor without associated KMP. The fairly rapid regrowth of this tumor, requiring adjuvant treatment after resection, is consistent with a potential for locally aggressive growth in a TA occurring in the brain.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/cirugía , Hemangioma/diagnóstico por imagen , Hemangioma/cirugía , Procedimientos Neuroquirúrgicos/métodos , Radiocirugia/métodos , Neoplasias Cutáneas/diagnóstico por imagen , Neoplasias Cutáneas/cirugía , Anciano , Edema Encefálico/etiología , Capilares/diagnóstico por imagen , Humanos , Síndrome de Kasabach-Merritt/diagnóstico por imagen , Síndrome de Kasabach-Merritt/cirugía , Masculino , Recurrencia Local de Neoplasia , Resultado del Tratamiento
10.
Stem Cell Res ; 16(3): 776-81, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27153350

RESUMEN

Reprogramming technology enables the production of neural progenitor cells (NPCs) from somatic cells by direct transdifferentiation. However, little is known on how neural programs in these induced neural stem cells (iNSCs) differ from those of alternative stem cell populations in vitro and in vivo. Here, we performed transcriptome analyses on murine iNSCs in comparison to brain-derived neural stem cells (NSCs) and pluripotent stem cell-derived NPCs, which revealed distinct global, neural, metabolic and cell cycle-associated marks in these populations. iNSCs carried a hindbrain/posterior cell identity, which could be shifted towards caudal, partially to rostral but not towards ventral fates in vitro. iNSCs survived after transplantation into the rodent brain and exhibited in vivo-characteristics, neural and metabolic programs similar to transplanted NSCs. However, iNSCs vastly retained caudal identities demonstrating cell-autonomy of regional programs in vivo. These data could have significant implications for a variety of in vitro- and in vivo-applications using iNSCs.


Asunto(s)
Encéfalo/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células-Madre Neurales/metabolismo , Transcriptoma , Animales , Encéfalo/patología , Diferenciación Celular , Células Cultivadas , Reprogramación Celular , Análisis por Conglomerados , Fibroblastos/citología , Perfilación de la Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/trasplante , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
Haematologica ; 100(1): 32-41, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25326431

RESUMEN

Epigenetic memory in induced pluripotent stem cells, which is related to the somatic cell type of origin of the stem cells, might lead to variations in the differentiation capacities of the pluripotent stem cells. In this context, induced pluripotent stem cells from human CD34(+) hematopoietic stem cells might be more suitable for hematopoietic differentiation than the commonly used fibroblast-derived induced pluripotent stem cells. To investigate the influence of an epigenetic memory on the ex vivo expansion of induced pluripotent stem cells into erythroid cells, we compared induced pluripotent stem cells from human neural stem cells and human cord blood-derived CD34(+) hematopoietic stem cells and evaluated their potential for differentiation into hematopoietic progenitor and mature red blood cells. Although genome-wide DNA methylation profiling at all promoter regions demonstrates that the epigenetic memory of induced pluripotent stem cells is influenced by the somatic cell type of origin of the stem cells, we found a similar hematopoietic induction potential and erythroid differentiation pattern of induced pluripotent stem cells of different somatic cell origin. All human induced pluripotent stem cell lines showed terminal maturation into normoblasts and enucleated reticulocytes, producing predominantly fetal hemoglobin. Differences were only observed in the growth rate of erythroid cells, which was slightly higher in the induced pluripotent stem cells derived from CD34(+) hematopoietic stem cells. More detailed methylation analysis of the hematopoietic and erythroid promoters identified similar CpG methylation levels in the induced pluripotent stem cell lines derived from CD34(+) cells and those derived from neural stem cells, which confirms their comparable erythroid differentiation potential.


Asunto(s)
Diferenciación Celular , Células Eritroides/citología , Sangre Fetal/citología , Células Madre Hematopoyéticas/citología , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Biomarcadores/metabolismo , Metilación de ADN , Epigenómica , Células Eritroides/metabolismo , Sangre Fetal/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células-Madre Neurales/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Cell Rep ; 8(6): 1697-1703, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25220454

RESUMEN

The differentiation capability of induced pluripotent stem cells (iPSCs) toward certain cell types for disease modeling and drug screening assays might be influenced by their somatic cell of origin. Here, we have compared the neural induction of human iPSCs generated from fetal neural stem cells (fNSCs), dermal fibroblasts, or cord blood CD34(+) hematopoietic progenitor cells. Neural progenitor cells (NPCs) and neurons could be generated at similar efficiencies from all iPSCs. Transcriptomics analysis of the whole genome and of neural genes revealed a separation of neuroectoderm-derived iPSC-NPCs from mesoderm-derived iPSC-NPCs. Furthermore, we found genes that were similarly expressed in fNSCs and neuroectoderm, but not in mesoderm-derived iPSC-NPCs. Notably, these neural signatures were retained after transplantation into the cortex of mice and paralleled with increased survival of neuroectoderm-derived cells in vivo. These results indicate distinct origin-dependent neural cell identities in differentiated human iPSCs both in vitro and in vivo.


Asunto(s)
Encéfalo/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Animales , Antígenos CD34/metabolismo , Diferenciación Celular , Células Cultivadas , Sangre Fetal/citología , Sangre Fetal/metabolismo , Feto/citología , Fibroblastos/citología , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Hematopoyéticas/citología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Mesodermo/citología , Ratones , Ratones Endogámicos NOD , Microscopía Confocal , Placa Neural/citología
14.
Neuropathology ; 34(2): 159-63, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23889676

RESUMEN

Gangliogliomas are well-differentiated, mixed glio-neuronal tumors of the CNS that are most frequently localized within the temporal lobe. In a minority of cases, gangliogliomas have been described in the brain stem where they may critically impinge anatomical structures. Rarely, ganglioglioma develop in cranial nerves, almost exclusively in the optic pathway, where they usually present as singular space-occupying masses. Here, we report on an 83-year-old patient who presented with unusual symmetrical, bilateral gangliogliomas of the trigeminal nerves. These tumors showed an exophytic growth within the subarachnoid space toward the Gasserian ganglion and surprisingly appeared as isointense masses on T1- and T2-weighted MRI. Due to their bilateral appearance, we performed array-comparative genomic hybridization (aCGH) on the gangliogliomas to address the possibility of an underlying tumor syndrome in this patient. To our knowledge, this is the first case of bilateral ganglioglioma of the trigeminal nerve described so far.


Asunto(s)
Neoplasias de los Nervios Craneales/patología , Ganglioglioma/patología , Enfermedades del Nervio Trigémino/patología , Anciano de 80 o más Años , Encéfalo/patología , Neoplasias de los Nervios Craneales/genética , Demencia/etiología , Ganglioglioma/genética , Humanos , Inmunohistoquímica , Masculino , Nervio Trigémino/patología , Enfermedades del Nervio Trigémino/genética
15.
J Neurol Surg A Cent Eur Neurosurg ; 74 Suppl 1: e203-6, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23670807

RESUMEN

BACKGROUND: Low-grade gliomas (e.g., pilocytic astrocytomas) are frequently found in patients with neurofibromatosis type 1 (NF1). Whereas most of those lesions are located supratentorially, cerebellar manifestations are described in < 1%. Malignant variants like glioblastoma and anaplastic astrocytoma (AA) are only rarely observed in NF1 patients. Thus, cerebellar AA is very infrequent and has not yet been described in an adult NF1 patient. CLINICAL PRESENTATION: We present the case of a 54-year-old male patient with von Recklinghausen disease who had a diffuse contrast-enhancing cerebellar mass that was resected guided by aminolevulinic acid (ALA)-fluorescence. Histopathological analyses revealed an AA with lack of pilocytic features or O6-methylguanine-DNA methyltransferase (MGMT) promoter hypermethylation. Due to the proximity of the tumor to the brainstem, adjuvant temozolomide chemotherapy was administered rather than first-line radiotherapy. Although the patient recovered quickly after the operation and tumor progression was ruled out in follow-up magnetic resonance imaging (MRI), the patient strongly deteriorated during a 16-month follow-up, and MRI revealed severe leukoencephalopathy. Extensive electrophysiological and radiological examination revealed a neurodegenerative disease of unknown etiology. Finally, the patient's condition improved receiving levodopa. CONCLUSIONS: A literature search yielded only one previously published case of an AA in a 9-year-old girl with NF1. Tumor control after resection was achieved in both patients; however, the patient in the mentioned report received radiation instead of temozolomide. In spite of different adjuvant therapies, tumor control for at least 16 months was achieved in both published cases. Thus, even though the role of adjuvant treatment options remains to be further elucidated, surgery is the appropriate therapy in these uncommon tumors providing mass reduction and histological diagnosis as well as tumor control.


Asunto(s)
Astrocitoma/cirugía , Neoplasias Cerebelosas/cirugía , Neurofibromatosis 1/complicaciones , Procedimientos Neuroquirúrgicos/métodos , Cirugía Asistida por Computador/métodos , Antineoplásicos Alquilantes/uso terapéutico , Astrocitoma/complicaciones , Astrocitoma/diagnóstico , Neoplasias Cerebelosas/complicaciones , Neoplasias Cerebelosas/diagnóstico , Terapia Combinada , Dacarbazina/análogos & derivados , Dacarbazina/uso terapéutico , Dopaminérgicos/uso terapéutico , Electrodiagnóstico , Electroencefalografía , Humanos , Levodopa/uso terapéutico , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Tomografía de Emisión de Positrones , Temozolomida , Tomografía Computarizada por Rayos X , Resultado del Tratamiento
16.
Curr Protoc Stem Cell Biol ; Chapter 2: Unit 2D.11, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22605646

RESUMEN

In vitro-differentiated embryonic stem (ES) cells comprise a useful source for cell replacement therapy, but the efficiency and safety of a translational approach are highly dependent on optimized protocols for directed differentiation of ES cells into the desired cell types in vitro. Furthermore, the transplantation of three-dimensional ES cell-derived structures instead of a single-cell suspension may improve graft survival and function by providing a beneficial microenvironment for implanted cells. To this end, we have developed a new method to efficiently differentiate mouse ES cells into neural aggregates that consist predominantly (>90%) of postmitotic neurons, neural progenitor cells, and radial glia-like cells. When transplanted into the excitotoxically lesioned striatum of adult mice, these substrate-adherent embryonic stem cell-derived neural aggregates (SENAs) showed significant advantages over transplanted single-cell suspensions of ES cell-derived neural cells, including improved survival of GABAergic neurons, increased cell migration, and significantly decreased risk of teratoma formation. Furthermore, SENAs mediated functional improvement after transplantation into animal models of Parkinson's disease and spinal cord injury. This unit describes in detail how SENAs are efficiently derived from mouse ES cells in vitro and how SENAs are isolated for transplantation. Furthermore, methods are presented for successful implantation of SENAs into animal models of Huntington's disease, Parkinson's disease, and spinal cord injury to study the effects of stem cell-derived neural aggregates in a disease context in vivo.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Embrionarias/citología , Células Madre Embrionarias/trasplante , Enfermedades del Sistema Nervioso/terapia , Neuronas/citología , Neuronas/trasplante , Trasplante de Células Madre , Animales , Adhesión Celular , Agregación Celular , Diferenciación Celular , Separación Celular , Modelos Animales de Enfermedad , Cuerpos Embrioides/citología , Femenino , Enfermedad de Huntington/terapia , Ratones , Ratones Endogámicos C57BL , Enfermedad de Parkinson/terapia , Traumatismos de la Médula Espinal/terapia
17.
PLoS One ; 6(5): e19926, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21655272

RESUMEN

Neural stem cells (NSCs) lose their competency to generate region-specific neuronal populations at an early stage during embryonic brain development. Here we investigated whether epigenetic modifications can reverse the regional restriction of mouse adult brain subventricular zone (SVZ) NSCs. Using a variety of chemicals that interfere with DNA methylation and histone acetylation, we showed that such epigenetic modifications increased neuronal differentiation but did not enable specific regional patterning, such as midbrain dopaminergic (DA) neuron generation. Only after Oct-4 overexpression did adult NSCs acquire a pluripotent state that allowed differentiation into midbrain DA neurons. DA neurons derived from Oct4-reprogrammed NSCs improved behavioural motor deficits in a rat model of Parkinson's disease (PD) upon intrastriatal transplantation. Here we report for the first time the successful differentiation of SVZ adult NSCs into functional region-specific midbrain DA neurons, by means of Oct-4 induced pluripotency.


Asunto(s)
Diferenciación Celular/fisiología , Reprogramación Celular , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Animales , Western Blotting , Diferenciación Celular/genética , Células Cultivadas , Dopamina/metabolismo , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Células Madre Pluripotentes Inducidas/trasplante , Ratones , Neuronas/citología , Neuronas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/terapia , Reacción en Cadena de la Polimerasa , Ratas , Ratas Sprague-Dawley
18.
J Neurotrauma ; 28(9): 1921-37, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21671795

RESUMEN

A major obstacle for the transplantation of neural stem cells (NSCs) into the lesioned spinal cord is their predominant astrocytic differentiation after transplantation. We took advantage of this predominant astrocytic differentiation of NSCs and expressed the paradigmatic beneficial neural cell adhesion molecule L1 in radial glial cells and reactive and nonreactive astrocytes as novel cellular vehicles to express L1 under the control of the promoter for the human glial fibrillary acidic protein (GFAP-L1 NSCs). Behavioral analysis and electrophysiological H-reflex recordings revealed that mice transplanted with GFAP-L1 NSCs showed enhanced locomotor recovery in comparison to mice injected with wild type (WT) NSCs or control mice injected with phosphate-buffered saline (PBS). This functional recovery was further accelerated in mice transplanted with L1-expressing radial glial cells that had been immunoisolated from GFAP-L1 NSCs (GFAP-L1-i cells). Morphological analysis revealed that mice grafted with GFAP-L1 NSCs exhibited increased neuronal differentiation and migration of transplanted cells, as well as increased soma size and cholinergic synaptic coverage of host motoneurons and increased numbers of endogenous catecholaminergic nerve fibers caudal to the lesion site. These findings show that L1-expressing astrocytes and radial glial cells isolated from GFAP-L1 NSC cultures represent a novel strategy for improving functional recovery after spinal cord injury, encouraging the use of the human GFAP promoter to target beneficial transgene expression in transplanted stem cells.


Asunto(s)
Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Células-Madre Neurales/trasplante , Neuroglía/trasplante , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/terapia , Animales , Movimiento Celular , Proliferación Celular , Femenino , Gliosis/metabolismo , Ratones , Actividad Motora/fisiología , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Neuroglía/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/fisiopatología
19.
Stem Cells ; 29(7): 1052-63, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21608081

RESUMEN

Immune rejection and risk of tumor formation are perhaps the greatest hurdles in the field of stem cell transplantation. Here, we report the generation of several lines of induced pluripotent stem cells (iPSCs) from cynomolgus macaque (CM) skin fibroblasts carrying specific major histocompatibility complex (MHC) haplotypes. To develop a collection of MHC-matched iPSCs, we genotyped the MHC locus of 25 CMs by microsatellite polymerase chain reaction analysis. Using retroviral infection of dermal skin fibroblasts, we generated several CM-iPSC lines carrying different haplotypes. We characterized the immunological properties of CM-iPSCs and demonstrated that CM-iPSCs can be induced to differentiate in vitro along specific neuronal populations, such as midbrain dopaminergic (DA) neurons. Midbrain-like DA neurons generated from CM-iPSCs integrated into the striatum of a rodent model of Parkinson's disease and promoted behavioral recovery. Importantly, neither tumor formation nor inflammatory reactions were observed in the transplanted animals up to 6 months after transplantation. We believe that the generation and characterization of such histocompatible iPSCs will allow the preclinical validation of safety and efficacy of iPSCs for neurodegenerative diseases and several other human conditions in the field of regenerative medicine.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/trasplante , Neuronas/trasplante , Trasplante de Células Madre/métodos , Animales , Diferenciación Celular/fisiología , Cuerpo Estriado/cirugía , Femenino , Humanos , Células Madre Pluripotentes Inducidas/inmunología , Macaca fascicularis , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neuronas/inmunología , Enfermedad de Parkinson/inmunología , Enfermedad de Parkinson/cirugía , Ratas , Ratas Sprague-Dawley
20.
PLoS One ; 6(3): e17126, 2011 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-21445247

RESUMEN

An obstacle to early stem cell transplantation into the acutely injured spinal cord is poor survival of transplanted cells. Transplantation of embryonic stem cells as substrate adherent embryonic stem cell-derived neural aggregates (SENAs) consisting mainly of neurons and radial glial cells has been shown to enhance survival of grafted cells in the injured mouse brain. In the attempt to promote the beneficial function of these SENAs, murine embryonic stem cells constitutively overexpressing the neural cell adhesion molecule L1 which favors axonal growth and survival of grafted and imperiled cells in the inhibitory environment of the adult mammalian central nervous system were differentiated into SENAs and transplanted into the spinal cord three days after compression lesion. Mice transplanted with L1 overexpressing SENAs showed improved locomotor function when compared to mice injected with wild-type SENAs. L1 overexpressing SENAs showed an increased number of surviving cells, enhanced neuronal differentiation and reduced glial differentiation after transplantation when compared to SENAs not engineered to overexpress L1. Furthermore, L1 overexpressing SENAs rescued imperiled host motoneurons and parvalbumin-positive interneurons and increased numbers of catecholaminergic nerve fibers distal to the lesion. In addition to encouraging the use of embryonic stem cells for early therapy after spinal cord injury L1 overexpression in the microenvironment of the lesioned spinal cord is a novel finding in its functions that would make it more attractive for pre-clinical studies in spinal cord regeneration and most likely other diseases of the nervous system.


Asunto(s)
Células Madre Embrionarias/metabolismo , Interneuronas/citología , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Traumatismos de la Médula Espinal/rehabilitación , Animales , Astrocitos/citología , Diferenciación Celular , Ratones , Neuroglía/citología , Traumatismos de la Médula Espinal/metabolismo , Trasplante de Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA