Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 209
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(29): e2110746119, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35858361

RESUMEN

Intraneuronal inclusions of misfolded α-synuclein (α-syn) and prion-like spread of the pathologic α-syn contribute to progressive neuronal death in Parkinson's disease (PD). Despite the pathologic significance, no efficient therapeutic intervention targeting α-synucleinopathy has been developed. In this study, we provide evidence that astrocytes, especially those cultured from the ventral midbrain (VM), show therapeutic potential to alleviate α-syn pathology in multiple in vitro and in vivo α-synucleinopathic models. Regulation of neuronal α-syn proteostasis underlies the therapeutic function of astrocytes. Specifically, VM-derived astrocytes inhibited neuronal α-syn aggregation and transmission in a paracrine manner by correcting not only intraneuronal oxidative and mitochondrial stresses but also extracellular inflammatory environments, in which α-syn proteins are prone to pathologic misfolding. The astrocyte-derived paracrine factors also promoted disassembly of extracellular α-syn aggregates. In addition to the aggregated form of α-syn, VM astrocytes reduced total α-syn protein loads both by actively scavenging extracellular α-syn fibrils and by a paracrine stimulation of neuronal autophagic clearance of α-syn. Transplantation of VM astrocytes into the midbrain of PD model mice alleviated α-syn pathology and protected the midbrain dopamine neurons from neurodegeneration. We further showed that cografting of VM astrocytes could be exploited in stem cell-based therapy for PD, in which host-to-graft transmission of α-syn pathology remains a critical concern for long-term cell therapeutic effects.


Asunto(s)
Astrocitos , Trasplante de Tejido Encefálico , Enfermedad de Parkinson , Proteostasis , alfa-Sinucleína , Animales , Astrocitos/trasplante , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Mesencéfalo/patología , Mesencéfalo/cirugía , Ratones , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/terapia , alfa-Sinucleína/metabolismo
2.
Acta Neuropathol ; 142(1): 117-137, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33913039

RESUMEN

Loss-of-function variants in the low-density lipoprotein receptor-related protein 10 (LRP10) gene have been associated with autosomal-dominant Parkinson's disease (PD), PD dementia, and dementia with Lewy bodies (DLB). Moreover, LRP10 variants have been found in individuals diagnosed with progressive supranuclear palsy and amyotrophic lateral sclerosis. Despite this genetic evidence, little is known about the expression and function of LRP10 protein in the human brain under physiological or pathological conditions. To better understand how LRP10 variants lead to neurodegeneration, we first performed an in-depth characterisation of LRP10 expression in post-mortem brains and human-induced pluripotent stem cell (iPSC)-derived astrocytes and neurons from control subjects. In adult human brain, LRP10 is mainly expressed in astrocytes and neurovasculature but undetectable in neurons. Similarly, LRP10 is highly expressed in iPSC-derived astrocytes but cannot be observed in iPSC-derived neurons. In astrocytes, LRP10 is present at trans-Golgi network, plasma membrane, retromer, and early endosomes. Interestingly, LRP10 also partially co-localises and interacts with sortilin-related receptor 1 (SORL1). Furthermore, although LRP10 expression and localisation in the substantia nigra of most idiopathic PD and DLB patients and LRP10 variant carriers diagnosed with PD or DLB appeared unchanged compared to control subjects, significantly enlarged LRP10-positive vesicles were detected in a patient carrying the LRP10 p.Arg235Cys variant. Last, LRP10 was detected in Lewy bodies (LB) at late maturation stages in brains from idiopathic PD and DLB patients and in LRP10 variant carriers. In conclusion, high LRP10 expression in non-neuronal cells and undetectable levels in neurons of control subjects indicate that LRP10-mediated pathogenicity is initiated via cell non-autonomous mechanisms, potentially involving the interaction of LRP10 with SORL1 in vesicle trafficking pathways. Together with the specific pattern of LRP10 incorporation into mature LBs, these data support an important mechanistic role for disturbed vesicle trafficking and loss of LRP10 function in neurodegenerative diseases.


Asunto(s)
Encéfalo/metabolismo , Proteínas Relacionadas con Receptor de LDL/genética , Cuerpos de Lewy/metabolismo , Enfermedad por Cuerpos de Lewy/metabolismo , Proteínas de Transporte de Membrana/genética , Enfermedad de Parkinson/metabolismo , Adulto , Anciano , Astrocitos/metabolismo , Astrocitos/trasplante , Encéfalo/citología , Encéfalo/patología , Variación Genética , Humanos , Células Madre Pluripotentes Inducidas/trasplante , Cuerpos de Lewy/patología , Enfermedad por Cuerpos de Lewy/patología , Persona de Mediana Edad , Enfermedades Neurodegenerativas/patología , Neuronas/trasplante , Enfermedad de Parkinson/patología
3.
Mol Cell Biochem ; 476(2): 619-631, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33070275

RESUMEN

Parkinson's disease (PD) is a disorder characterized by a progressive loss of the dopaminergic neurons in the substantia nigra and a depletion of the neurotransmitter dopamine in the striatum. Our published results indicate that fasciculation and elongation protein zeta-1 (FEZ1) plays a role in the astrocyte-mediated protection of dopamine neurons and regulation of the neuronal microenvironment during the progression of PD. In this study, we examined the effects of engrafted type-2 astrocytes (T2As) with high expression of FEZ1 on the improvement of the symptoms and functional reconstruction of PD rats. T2As were stereotactically transplanted into the striatum of rats with PD induced by 6-hydroxydopamine (6-OHDA). An examination of apomorphine (APO)-induced rotations was performed to evaluate dopamine neuron damage and motor functions. Remarkably, the grafted cells survived in the lesion environment for six weeks or longer after implantation. In addition, the transplantation of T2As decrease the average velocity and the duration time of the APO-induced rotations, and increase the actuation time, as measured in the rotation behavioural tests. In the substantia nigra, the transplantation of T2As reduced the PD-induced GFAP, TH and FEZ1 downregulation. The grafted cells exclusively migrated to other regions near the injection site in the striatum and differentiated into GFAP+ astrocytes or TH+ neurons. Furthermore, by detecting monoamine neurotransmitters through high-performance liquid chromatography, we found that the nigrostriatal pathway had been repaired to some extent. Taken together, these results suggest that engrafted T2As with high expression of FEZ1 improved the symptoms and functional reconstruction of PD rats, providing a theoretical basis for FEZ1 as a potential target and engraftment of T2As as a therapeutic strategy in the treatment of PD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apomorfina/farmacología , Astrocitos/trasplante , Neuronas Dopaminérgicas/efectos de los fármacos , Enfermedad de Parkinson/terapia , Sustancia Negra/metabolismo , Adrenérgicos/administración & dosificación , Animales , Astrocitos/citología , Astrocitos/metabolismo , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Masculino , Actividad Motora/efectos de los fármacos , Oxidopamina/administración & dosificación , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Ratas , Ratas Sprague-Dawley
4.
Int J Mol Sci ; 22(22)2021 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-34830023

RESUMEN

Transplantation of exogenous dopaminergic (DA) neurons is an alternative strategy to replenish DA neurons that have lost along the course of Parkinson's disease (PD). From the perspective of ethical acceptation, the source limitations, and the intrinsic features of PD pathology, astrocytes (AS) and mesenchymal stem cells (MSCs) are the two promising candidates of DA induction. In the present study, we induced AS or MSCs primary culture by the combination of the classical transcription-factor cocktails Mash1, Lmx1a, and Nurr1 (MLN), the chemical cocktails (S/C/D), and the morphogens SHH, FGF8, and FGF2 (S/F8/F2); the efficiency of induction into DA neurons was further analyzed by using immunostaining against the DA neuronal markers. AS could be efficiently converted into the DA neurons in vitro by the transcriptional regulation of MLN, and the combination with S/C/D or S/F8/F2 further increased the conversion efficiency. In contrast, MSCs from umbilical cord (UC-MSCs) or adipose tissue (AD-MSCs) showed moderate TH immunoreactivity after the induction with S/F8/F2 instead of with MLN or S/C/D. Our data demonstrated that AS and MSCs held lineage-specific molecular codes on the induction into DA neurons and highlighted the unique superiority of AS in the potential of cell replacement therapy for PD.


Asunto(s)
Astrocitos/trasplante , Neuronas Dopaminérgicas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Enfermedad de Parkinson/terapia , Animales , Astrocitos/metabolismo , Diferenciación Celular/genética , Dopamina/metabolismo , Neuronas Dopaminérgicas/patología , Neuronas Dopaminérgicas/trasplante , Humanos , Trasplante de Células Madre Mesenquimatosas , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Cultivo Primario de Células , Ratas , Factores de Transcripción/genética , Cordón Umbilical/metabolismo , Cordón Umbilical/trasplante
5.
Stem Cells ; 37(10): 1344-1356, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31287930

RESUMEN

Due to the limited capacity of brain tissue to self-regenerate after traumatic brain injury (TBI), the mobilization of endogenous neural stem cells (NSCs) is a popular research topic. In the clinic, the neurogenic abilities of adults versus neonates vary greatly, which is likely related to functional differences in NSCs. Recent studies have demonstrated that the molecules secreted from astrocytes play important roles in NSC fate determination. In this study, conditioned media (CM) derived from neonatal or adult rat astrocytes, which were unstimulated or stimulated by lipopolysaccharide (LPS), were prepared to treat NSCs. Our results revealed that neonatal rat astrocytes can significantly promote the proliferation of NSCs, compared with adult rat astrocytes, regardless of whether or not they were stimulated by LPS. Furthermore, we used mass spectrometry to detect the constituents of the CM from each group. We analyzed and screened for a protein, Tenascin-C (TNC), which was highly expressed in the neonatal group but poorly expressed in the adult group. We found that TNC can bind to the NSC surface epidermal growth factor receptor and promote proliferation through the PI3K-AKT pathway in vitro. Additionally, we confirmed in vivo that TNC can promote damage repair in a rat model of TBI, through enhancing the proliferation of endogenous NSCs. We believe that these findings provide a mechanistic understanding of why neonates show better neuroregenerative abilities than adults. This also provides a potential future therapeutic target, TNC, for injury repair after TBI. Stem Cells 2019;37:1344-1356.


Asunto(s)
Astrocitos/trasplante , Lesiones Traumáticas del Encéfalo/terapia , Células-Madre Neurales/trasplante , Animales , Diferenciación Celular , Proliferación Celular , Humanos , Ratas
7.
Nanomedicine ; 21: 102048, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31271878

RESUMEN

Neurodegenerative diseases and traumatic brain injuries can destroy neurons, resulting in sensory and motor function loss. Transplantation of differentiated neurons from stem cells could help restore such lost functions. Plasmonic gold nanorods (AuNR) were integrated in growth surfaces to stimulate and modulate neural cells in order to tune cell physiology. An AuNR nanocomposite system was fabricated, characterized, and then utilized to study the differentiation of embryonic rat neural stem cells (NSCs). Results demonstrated that this plasmonic surface 1) accelerated differentiation, yielding almost twice as many differentiated neural cells as a traditional NSC culture surface coated with poly-D-lysine and laminin for the same time period; and 2) promoted differentiation of NSCs into neurons and astrocytes in a 2:1 ratio, as evidenced by the expression of relevant marker proteins. These results indicate that the design and properties of this AuNR plasmonic surface would be advantageous for tissue engineering to address neural degeneration.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Nanotubos/química , Enfermedades Neurodegenerativas/terapia , Neuronas/trasplante , Animales , Astrocitos/trasplante , Lesiones Traumáticas del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/terapia , Células Cultivadas , Células Madre Embrionarias/efectos de los fármacos , Oro/química , Oro/farmacología , Humanos , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/trasplante , Enfermedades Neurodegenerativas/patología , Neuronas/efectos de los fármacos , Ratas
8.
Glia ; 65(12): 1867-1884, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28631858

RESUMEN

Rapid advances in Ca2+ imaging techniques enable us to simultaneously monitor the activities of hundreds of astrocytes in the intact brain, thus providing a powerful tool for understanding the functions of both host and engrafted astrocytes in sensory processing in vivo. These techniques include both improved Ca2+ indicators and advanced optical recording methods. Astrocytes in multiple cortical and sub-cortical areas are able to respond to the corresponding sensory modalities. These sensory stimuli produce astrocytic Ca2+ responses through different cellular mechanisms. In addition, it has been suggested that astrocytic gene deficiencies in various sensory systems cause impairments in sensory circuits and cognition. Therefore, glial transplantation would be a potentially interesting approach for the cell-based therapy for glia-related disorders. There are multiple cell sources for glial transplantation, including neural stem cells, glial progenitors, and pluripotent stem cells. Both in vitro and in vivo studies have shown that engrafted astrocytes derived from these cell sources are capable of responding to sensory stimulation by elevating the intracellular Ca2+ concentration. These results indicate that engrafted astrocytes not only morphologically but also functionally integrate into the host neural network. Until now, many animal studies have proven that glial transplantation would be a good choice for treating multiple glial disorders. Together, these studies on the sensory responses of host and engrafted astrocytes have provided us a novel perspective in both neuron-glia circuit functions and future treatment strategies for glial disorders.


Asunto(s)
Vías Aferentes/fisiología , Astrocitos/fisiología , Astrocitos/trasplante , Encéfalo/citología , Sensación/fisiología , Animales , Calcio/metabolismo , Humanos , Células Madre/fisiología
9.
Cereb Cortex ; 26(9): 3690-3704, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27405333

RESUMEN

Glial precursor transplantation provides a potential therapy for brain disorders. Before its clinical application, experimental evidence needs to indicate that engrafted glial cells are functionally incorporated into the existing circuits and become essential partners of neurons for executing fundamental brain functions. While previous experiments supporting for their functional integration have been obtained under in vitro conditions using slice preparations, in vivo evidence for such integration is still lacking. Here, we utilized in vivo two-photon Ca(2+) imaging along with immunohistochemistry, fluorescent indicator labeling-based axon tracing and correlated light/electron microscopy to analyze the profiles and the functional status of glial precursor cell-derived astrocytes in adult mouse neocortex. We show that after being transplanted into somatosensory cortex, precursor-derived astrocytes are able to survive for more than a year and respond with Ca(2+) signals to sensory stimulation. These sensory-evoked responses are mediated by functionally-expressed nicotinic receptors and newly-established synaptic contacts with the host cholinergic afferents. Our results provide in vivo evidence for a functional integration of transplanted astrocytes into adult mammalian neocortex, representing a proof-of-principle for sensory cortex remodeling through addition of essential neural elements. Moreover, we provide strong support for the use of glial precursor transplantation to understand glia-related neural development in vivo.


Asunto(s)
Astrocitos/fisiología , Astrocitos/trasplante , Señalización del Calcio/fisiología , Estimulación Eléctrica/métodos , Neocórtex/citología , Neocórtex/fisiología , Corteza Somatosensorial/fisiología , Potenciales de Acción/fisiología , Animales , Astrocitos/citología , Diferenciación Celular/fisiología , Femenino , Humanos , Masculino , Ratones , Neocórtex/cirugía , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Células-Madre Neurales/trasplante , Transducción de Señal/fisiología
10.
Glia ; 64(1): 63-75, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26295203

RESUMEN

Astrocytes are instrumental to major brain functions, including metabolic support, extracellular ion regulation, the shaping of excitatory signaling events and maintenance of synaptic glutamate homeostasis. Astrocyte dysfunction contributes to numerous developmental, psychiatric and neurodegenerative disorders. The generation of adult human fibroblast-derived induced pluripotent stem cells (iPSCs) has provided novel opportunities to study mechanisms of astrocyte dysfunction in human-derived cells. To overcome the difficulties of cell type heterogeneity during the differentiation process from iPSCs to astroglial cells (iPS astrocytes), we generated homogenous populations of iPS astrocytes using zinc-finger nuclease (ZFN) technology. Enhanced green fluorescent protein (eGFP) driven by the astrocyte-specific glial fibrillary acidic protein (GFAP) promoter was inserted into the safe harbor adeno-associated virus integration site 1 (AAVS1) locus in disease and control-derived iPSCs. Astrocyte populations were enriched using Fluorescence Activated Cell Sorting (FACS) and after enrichment more than 99% of iPS astrocytes expressed mature astrocyte markers including GFAP, S100ß, NFIA and ALDH1L1. In addition, mature pure GFP-iPS astrocytes exhibited a well-described functional astrocytic activity in vitro characterized by neuron-dependent regulation of glutamate transporters to regulate extracellular glutamate concentrations. Engraftment of GFP-iPS astrocytes into rat spinal cord grey matter confirmed in vivo cell survival and continued astrocytic maturation. In conclusion, the generation of GFAP::GFP-iPS astrocytes provides a powerful in vitro and in vivo tool for studying astrocyte biology and astrocyte-driven disease pathogenesis and therapy.


Asunto(s)
Astrocitos/fisiología , Ingeniería Celular/métodos , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Animales , Astrocitos/trasplante , Supervivencia Celular/fisiología , Células Cultivadas , Desoxirribonucleasas , Dependovirus/genética , Fibroblastos/fisiología , Genes Reporteros , Vectores Genéticos , Sustancia Gris/citología , Sustancia Gris/fisiología , Sustancia Gris/cirugía , Proteínas Fluorescentes Verdes/genética , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Ratones , Regiones Promotoras Genéticas , Ratas Sprague-Dawley , Médula Espinal/citología , Médula Espinal/fisiología , Médula Espinal/cirugía , Dedos de Zinc
11.
Glia ; 64(1): 48-62, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26296243

RESUMEN

Fatty acid binding protein 7 (FABP7) expressed by astrocytes in developing and mature brains is involved in uptake and transportation of fatty acids, signal transduction, and gene transcription. Fabp7 knockout (Fabp7 KO) mice show behavioral phenotypes reminiscent of human neuropsychiatric disorders such as schizophrenia. However, direct evidence showing how FABP7 deficiency in astrocytes leads to altered brain function is lacking. Here, we examined neuronal dendritic morphology and synaptic plasticity in medial prefrontal cortex (mPFC) of Fabp7 KO mice and in primary cortical neuronal cultures. Golgi staining of cortical pyramidal neurons in Fabp7 KO mice revealed aberrant dendritic morphology and decreased spine density compared with those in wild-type (WT) mice. Aberrant dendritic morphology was also observed in primary cortical neurons co-cultured with FABP7-deficient astrocytes and neurons cultured in Fabp7 KO astrocyte-conditioned medium. Excitatory synapse number was decreased in mPFC of Fabp7 KO mice and in neurons co-cultured with Fabp7 KO astrocytes. Accordingly, whole-cell voltage-clamp recording in brain slices from pyramidal cells in the mPFC showed that both amplitude and frequency of action potential-independent miniature excitatory postsynaptic currents (mEPSCs) were decreased in Fabp7 KO mice. Moreover, transplantation of WT astrocytes into the mPFC of Fabp7 KO mice partially attenuated behavioral impairments. Collectively, these results suggest that astrocytic FABP7 is important for dendritic arbor growth, neuronal excitatory synapse formation, and synaptic transmission, and provide new insights linking FABP7, lipid homeostasis, and neuropsychiatric disorders, leading to novel therapeutic interventions.


Asunto(s)
Astrocitos/fisiología , Dendritas/fisiología , Proteínas de Unión a Ácidos Grasos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Corteza Prefrontal/fisiología , Células Piramidales/fisiología , Sinapsis/fisiología , Animales , Astrocitos/trasplante , Técnicas de Cocultivo , Potenciales Postsinápticos Excitadores/fisiología , Proteína de Unión a los Ácidos Grasos 7 , Proteínas de Unión a Ácidos Grasos/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Potenciales Postsinápticos Miniatura/fisiología , Actividad Motora/fisiología , Proteínas del Tejido Nervioso/genética , Corteza Prefrontal/citología , Corteza Prefrontal/cirugía , Células Piramidales/citología
12.
Stem Cells ; 33(4): 1036-41, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25728093

RESUMEN

Spinal cord injury (SCI) usually results in long lasting locomotor and sensory neuron degeneration below the injury. Astrocytes normally play a decisive role in mechanical and metabolic support of neurons, but in the spinal cord they cause injury, exerting well-known detrimental effects that contribute to glial scar formation and inhibition of axon outgrowth. Cell transplantation is considered a promising approach for replacing damaged cells and promoting neuroprotective and neuroregenerative repair, but the effects of the grafted cells on local tissue and the regenerative properties of endogenous neural stem cells in the injured spinal cord are largely unknown. During the last 2 decades cumulative evidence from diverse animal models has indicated that reactive astrocytes in synergy with transplanted cells could be beneficial for injury in multiple ways, including neuroprotection and axonal growth. In this review, we specifically focus on the dual opposing roles of reactive astrocytes in SCI and how they contribute to the creation of a permissive environment when combined with transplanted cells as the influential components for a local regenerative niche. Modulation of reactive astrocyte function might represent an extremely attractive new therapy to enhance the functional outcomes in patients.


Asunto(s)
Astrocitos/metabolismo , Astrocitos/trasplante , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/terapia , Trasplante de Células Madre/métodos , Animales , Humanos , Regeneración Nerviosa/fisiología , Células Madre/metabolismo
13.
Mol Ther ; 23(3): 533-48, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25492561

RESUMEN

Approximately half of traumatic spinal cord injury (SCI) cases affect cervical regions, resulting in chronic respiratory compromise. The majority of these injuries affect midcervical levels, the location of phrenic motor neurons (PMNs) that innervate the diaphragm. A valuable opportunity exists following SCI for preventing PMN loss that occurs during secondary degeneration. One of the primary causes of secondary injury is excitotoxicity due to dysregulation of extracellular glutamate homeostasis. Astrocytes express glutamate transporter 1 (GLT1), which is responsible for the majority of CNS glutamate clearance. Given our observations of GLT1 dysfunction post-SCI, we evaluated intraspinal transplantation of Glial-Restricted Precursors (GRPs)--a class of lineage-restricted astrocyte progenitors--into ventral horn following cervical hemicontusion as a novel strategy for reconstituting GLT1 function, preventing excitotoxicity and protecting PMNs in the acutely injured spinal cord. We find that unmodified transplants express low levels of GLT1 in the injured spinal cord. To enhance their therapeutic properties, we engineered GRPs with AAV8 to overexpress GLT1 only in astrocytes using the GFA2 promoter, resulting in significantly increased GLT1 protein expression and functional glutamate uptake following astrocyte differentiation in vitro and after transplantation into C4 hemicontusion. Compared to medium-only control and unmodified GRPs, GLT1-overexpressing transplants reduced lesion size, diaphragm denervation and diaphragm dysfunction. Our findings demonstrate transplantation-based replacement of astrocyte GLT1 is a promising approach for SCI.


Asunto(s)
Astrocitos/trasplante , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Diafragma/metabolismo , Transportador 2 de Aminoácidos Excitadores/genética , Traumatismos de la Médula Espinal/terapia , Médula Espinal/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Dependovirus/genética , Diafragma/patología , Modelos Animales de Enfermedad , Transportador 2 de Aminoácidos Excitadores/metabolismo , Femenino , Expresión Génica , Genes Reporteros , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Transgénicos , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Nervio Frénico/lesiones , Nervio Frénico/metabolismo , Nervio Frénico/patología , Ratas , Ratas Sprague-Dawley , Recuperación de la Función , Médula Espinal/patología , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Transgenes
14.
Proc Natl Acad Sci U S A ; 110(17): 7038-43, 2013 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-23530235

RESUMEN

Cellular reprogramming is a new and rapidly emerging field in which somatic cells can be turned into pluripotent stem cells or other somatic cell types simply by the expression of specific combinations of genes. By viral expression of neural fate determinants, it is possible to directly reprogram mouse and human fibroblasts into functional neurons, also known as induced neurons. The resulting cells are nonproliferating and present an alternative to induced pluripotent stem cells for obtaining patient- and disease-specific neurons to be used for disease modeling and for development of cell therapy. In addition, because the cells do not pass a stem cell intermediate, direct neural conversion has the potential to be performed in vivo. In this study, we show that transplanted human fibroblasts and human astrocytes, which are engineered to express inducible forms of neural reprogramming genes, convert into neurons when reprogramming genes are activated after transplantation. Using a transgenic mouse model to specifically direct expression of reprogramming genes to parenchymal astrocytes residing in the striatum, we also show that endogenous mouse astrocytes can be directly converted into neural nuclei (NeuN)-expressing neurons in situ. Taken together, our data provide proof of principle that direct neural conversion can take place in the adult rodent brain when using transplanted human cells or endogenous mouse cells as a starting cell for neural conversion.


Asunto(s)
Astrocitos/trasplante , Diferenciación Celular/fisiología , Reprogramación Celular/fisiología , Fibroblastos/trasplante , Neuronas/citología , Animales , Astrocitos/citología , Reprogramación Celular/efectos de los fármacos , Cuerpo Estriado/citología , Doxiciclina/farmacología , Fibroblastos/citología , Citometría de Flujo , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes , Humanos , Lentivirus , Ratones , Ratones Transgénicos , Neuronas/fisiología , Ratas , Ratas Sprague-Dawley
15.
J Neurosci ; 34(7): 2438-43, 2014 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-24523534

RESUMEN

Traumatic spinal cord injury (SCI) results in a cascade of tissue responses leading to cell death, axonal degeneration, and glial scar formation, exacerbating the already hostile environment and further inhibiting axon regeneration. Overcoming these inhibitory cues and promoting axonal regeneration is one of the primary targets in developing a cure for SCI. Previously, we demonstrated that transplantation of bone morphogenetic protein (BMP)-induced astrocytes derived from embryonic glial-restricted precursors (GDAs(BMP)) promotes extensive axonal growth and motor function recovery in a rodent spinal cord injury model. Here, we identify periostin (POSTN), a secreted protein, as a key component of GDA(BMP)-induced axonal regeneration. POSTN is highly expressed by GDAs(BMP) and the perturbation of POSTN expression by shRNA diminished GDA(BMP)-induced neurite extension in vitro. We also found that recombinant POSTN is sufficient to overcome the inhibitory effect of scar-associated molecules and promote neurite extension in vitro by signaling through focal adhesion kinase and Akt. Furthermore, transplantation of POSTN-deficient GDAs(BMP) into the injured rat spinal cord resulted in compromised axonal regeneration, indicating that POSTN plays an essential role in GDA(BMP)-mediated axonal regeneration. This finding reveals not only one of the major mechanisms underlying GDA(BMP)-dependent recovery from SCI, but also the potential of POSTN as a therapeutic agent for traumatic injury of the CNS.


Asunto(s)
Astrocitos/metabolismo , Astrocitos/trasplante , Moléculas de Adhesión Celular/metabolismo , Regeneración Nerviosa/fisiología , Traumatismos de la Médula Espinal/metabolismo , Animales , Axones/metabolismo , Diferenciación Celular/fisiología , Modelos Animales de Enfermedad , Células Madre Embrionarias/citología , Células-Madre Neurales/citología , Ratas , Ratas Sprague-Dawley
16.
Cell Mol Life Sci ; 71(6): 999-1015, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24100629

RESUMEN

Neurodegenerative disorders are characterized by the selective vulnerability and progressive loss of discrete neuronal populations. Non-neuronal cells appear to significantly contribute to neuronal loss in diseases such as amyotrophic lateral sclerosis (ALS), Parkinson, and Alzheimer's disease. In ALS, there is deterioration of motor neurons in the cortex, brainstem, and spinal cord, which control voluntary muscle groups. This results in muscle wasting, paralysis, and death. Neuroinflammation, characterized by the appearance of reactive astrocytes and microglia as well as macrophage and T-lymphocyte infiltration, appears to be highly involved in the disease pathogenesis, highlighting the involvement of non-neuronal cells in neurodegeneration. There appears to be cross-talk between motor neurons, astrocytes, and immune cells, including microglia and T-lymphocytes, which are subsequently activated. Currently, effective therapies for ALS are lacking; however, the non-cell autonomous nature of ALS may indicate potential therapeutic targets. Here, we review the mechanisms of action of astrocytes, microglia, and T-lymphocytes in the nervous system in health and during the pathogenesis of ALS. We also evaluate the therapeutic potential of these cellular populations, after transplantation into ALS patients and animal models of the disease, in modulating the environment surrounding motor neurons from pro-inflammatory to neuroprotective. We also thoroughly discuss the recent advances made in the field and caveats that need to be overcome for clinical translation of cell therapies aimed at modulating non-cell autonomous events to preserve remaining motor neurons in patients.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Astrocitos/trasplante , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Microglía/trasplante , Linfocitos T/trasplante , Esclerosis Amiotrófica Lateral/inmunología , Esclerosis Amiotrófica Lateral/patología , Animales , Astrocitos/metabolismo , Células COS , Chlorocebus aethiops , Modelos Animales de Enfermedad , Humanos , Inflamación/inmunología , Inflamación/terapia , Macrófagos/inmunología , Ratones , Microglía/metabolismo , Neuronas Motoras/metabolismo , Linfocitos T/inmunología
17.
Bioessays ; 35(10): 876-80, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23897758

RESUMEN

What is the biological basis for human cognition? Our understanding why human brains make us smarter than other animals is still in its infancy. In recent years, astrocytes have been shown to be indispensable for neuronal survival, growth, synapse formation, and synapse function. Now, in a new study from Maiken Nedergaard and Steven Goldman's groups (Han et al., 2013), human glia progenitor cells have been transplanted into mouse forebrains. These progenitors survived, migrated widely, and gave rise to astrocytes that displayed the characteristics of human astrocytes in the rodent host brains. Strikingly, the mice with transplanted human cells displayed improved long term potentiation (LTP) and learning, suggesting the potential importance of human astrocytes in the unique cognitive abilities of human brains. This landmark paper is an important first step toward future investigations of whether and how human astrocytes play a role in distinguishing the cognitive abilities of humans from those of other animals.


Asunto(s)
Astrocitos/citología , Astrocitos/trasplante , Animales , Astrocitos/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Cognición/fisiología , Humanos , Aprendizaje/fisiología , Potenciación a Largo Plazo/fisiología , Memoria/fisiología , Ratones , Trasplante de Células Madre , Células Madre/citología , Células Madre/metabolismo , Sinapsis/metabolismo
19.
Proc Natl Acad Sci U S A ; 107(28): 12704-9, 2010 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-20615974

RESUMEN

Various types of induced pluripotent stem (iPS) cells have been established by different methods, and each type exhibits different biological properties. Before iPS cell-based clinical applications can be initiated, detailed evaluations of the cells, including their differentiation potentials and tumorigenic activities in different contexts, should be investigated to establish their safety and effectiveness for cell transplantation therapies. Here we show the directed neural differentiation of murine iPS cells and examine their therapeutic potential in a mouse spinal cord injury (SCI) model. "Safe" iPS-derived neurospheres, which had been pre-evaluated as nontumorigenic by their transplantation into nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mouse brain, produced electrophysiologically functional neurons, astrocytes, and oligodendrocytes in vitro. Furthermore, when the safe iPS-derived neurospheres were transplanted into the spinal cord 9 d after contusive injury, they differentiated into all three neural lineages without forming teratomas or other tumors. They also participated in remyelination and induced the axonal regrowth of host 5HT(+) serotonergic fibers, promoting locomotor function recovery. However, the transplantation of iPS-derived neurospheres pre-evaluated as "unsafe" showed robust teratoma formation and sudden locomotor functional loss after functional recovery in the SCI model. These findings suggest that pre-evaluated safe iPS clone-derived neural stem/progenitor cells may be a promising cell source for transplantation therapy for SCI.


Asunto(s)
Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , Animales , Astrocitos/patología , Astrocitos/trasplante , Axones/patología , Axones/trasplante , Diferenciación Celular/fisiología , Trasplante de Células , Células Cultivadas , Femenino , Células Madre Pluripotentes Inducidas , Ratones , Ratones Endogámicos C57BL , Actividad Motora/fisiología , Neuronas/citología , Neuronas/patología , Neuronas/trasplante , Oligodendroglía/citología , Oligodendroglía/fisiología , Oligodendroglía/trasplante , Recuperación de la Función/fisiología , Regeneración , Médula Espinal/citología , Médula Espinal/cirugía , Médula Espinal/trasplante , Traumatismos de la Médula Espinal/cirugía , Células Madre/patología
20.
Pharm Res ; 29(5): 1328-43, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22134779

RESUMEN

PURPOSE: To develop Fe(3)O(4)-PEI-RITC magnetic nanoparticles with multimodal MRI-fluorescence imaging and transfection capability, for use in neural cell replacement therapies. METHODS: The Fe(3)O(4)-PEI-RITC MNPs were synthesised through a multi-step chemical grafting procedure: (i) Silanisation of MNPs with 3-iodopropyltrimethoxysilane; (ii) PEI coupling with iodopropyl groups on the MNP surface; and (iii) RITC binding onto the PEI coating. The cell labelling and transfection capabilities of these particles were evaluated in astrocytes derived from primary cultures. RESULTS: Fe(3)O(4)-PEI-RITC MNPs did not exert acute toxic effects in astrocytes (at ≤ 6 days). Cells showed rapid and extensive particle uptake with up to 100% cellular labelling observed by 24 h. MRI and microscopy studies demonstrate that the particles have potential for use in bimodal MR-fluorescence imaging. Additionally, the particles were capable of delivering plasmids encoding reporter protein (approximately 4 kb) to astrocytes, albeit with low efficiencies. CONCLUSIONS: Multifunctional Fe(3)O(4)-PEI-RITC MNPs were successfully prepared using a multi-step synthetic pathway, with the PEI and RITC chemically bound onto the MNP surface. Their combined MR-fluorescence imaging capabilities with additional potential for transfection applications can provide a powerful tool, after further development, for non-invasive cell tracking and gene transfer to neural transplant populations.


Asunto(s)
Trasplante de Células , Compuestos Férricos/química , Técnicas de Transferencia de Gen , Iminas/química , Magnetismo , Nanopartículas , Polietilenos/química , Animales , Astrocitos/metabolismo , Astrocitos/trasplante , Células Cultivadas , Inmunohistoquímica , Microscopía Fluorescente , Ratas , Ratas Sprague-Dawley , Espectroscopía Infrarroja por Transformada de Fourier , Propiedades de Superficie , Difracción de Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA