Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Data ; 10(1): 24, 2023 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-36631473

RESUMEN

The National Institute of Health (NIH) Library of integrated network-based cellular signatures (LINCS) program is premised on the generation of a publicly available data resource of cell-based biochemical responses or "signatures" to genetic or environmental perturbations. NeuroLINCS uses human inducible pluripotent stem cells (hiPSCs), derived from patients and healthy controls, and differentiated into motor neuron cell cultures. This multi-laboratory effort strives to establish i) robust multi-omic workflows for hiPSC and differentiated neuronal cultures, ii) public annotated data sets and iii) relevant and targetable biological pathways of spinal muscular atrophy (SMA) and amyotrophic lateral sclerosis (ALS). Here, we focus on the proteomics and the quality of the developed workflow of hiPSC lines from 6 individuals, though epigenomics and transcriptomics data are also publicly available. Known and commonly used markers representing 73 proteins were reproducibly quantified with consistent expression levels across all hiPSC lines. Data quality assessments, data levels and metadata of all 6 genetically diverse human iPSCs analysed by DIA-MS are parsable and available as a high-quality resource to the public.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Proteoma , Humanos , Neuronas Motoras , Proteoma/metabolismo , Proteómica
2.
Nat Med ; 28(9): 1813-1822, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36064599

RESUMEN

Amyotrophic lateral sclerosis (ALS) involves progressive motor neuron loss, leading to paralysis and death typically within 3-5 years of diagnosis. Dysfunctional astrocytes may contribute to disease and glial cell line-derived neurotrophic factor (GDNF) can be protective. Here we show that human neural progenitor cells transduced with GDNF (CNS10-NPC-GDNF) differentiated to astrocytes protected spinal motor neurons and were safe in animal models. CNS10-NPC-GDNF were transplanted unilaterally into the lumbar spinal cord of 18 ALS participants in a phase 1/2a study (NCT02943850). The primary endpoint of safety at 1 year was met, with no negative effect of the transplant on motor function in the treated leg compared with the untreated leg. Tissue analysis of 13 participants who died of disease progression showed graft survival and GDNF production. Benign neuromas near delivery sites were common incidental findings at post-mortem. This study shows that one administration of engineered neural progenitors can provide new support cells and GDNF delivery to the ALS patient spinal cord for up to 42 months post-transplantation.


Asunto(s)
Esclerosis Amiotrófica Lateral , Células-Madre Neurales , Esclerosis Amiotrófica Lateral/terapia , Animales , Modelos Animales de Enfermedad , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Humanos , Médula Espinal , Superóxido Dismutasa
3.
iScience ; 24(11): 103221, 2021 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-34746695

RESUMEN

Neurodegenerative diseases are challenging for systems biology because of the lack of reliable animal models or patient samples at early disease stages. Induced pluripotent stem cells (iPSCs) could address these challenges. We investigated DNA, RNA, epigenetics, and proteins in iPSC-derived motor neurons from patients with ALS carrying hexanucleotide expansions in C9ORF72. Using integrative computational methods combining all omics datasets, we identified novel and known dysregulated pathways. We used a C9ORF72 Drosophila model to distinguish pathways contributing to disease phenotypes from compensatory ones and confirmed alterations in some pathways in postmortem spinal cord tissue of patients with ALS. A different differentiation protocol was used to derive a separate set of C9ORF72 and control motor neurons. Many individual -omics differed by protocol, but some core dysregulated pathways were consistent. This strategy of analyzing patient-specific neurons provides disease-related outcomes with small numbers of heterogeneous lines and reduces variation from single-omics to elucidate network-based signatures.

4.
Stem Cell Reports ; 10(6): 1696-1704, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29706501

RESUMEN

Trophic factor delivery to the brain using stem cell-derived neural progenitors is a powerful way to bypass the blood-brain barrier. Protection of diseased neurons using this technology is a promising therapy for neurodegenerative diseases. Glial cell line-derived neurotrophic factor (GDNF) has provided benefits to Parkinsonian patients and is being used in a clinical trial for amyotrophic lateral sclerosis. However, chronic trophic factor delivery prohibits dose adjustment or cessation if side effects develop. To address this, we engineered a doxycycline-regulated vector, allowing inducible and reversible expression of a therapeutic molecule. Human induced pluripotent stem cell (iPSC)-derived neural progenitors were stably transfected with the vector and transplanted into the adult mouse brain. Doxycycline can penetrate the graft, with addition and withdrawal providing inducible and reversible GDNF expression in vivo, over multiple cycles. Our findings provide proof of concept for combining gene and stem cell therapy for effective modulation of ectopic protein expression in transplanted cells.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Trasplante de Células Madre , Tratamiento Basado en Trasplante de Células y Tejidos , Expresión Génica , Genes Reporteros , Terapia Genética , Vectores Genéticos/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Plantas Modificadas Genéticamente , Trasplante de Células Madre/métodos , Transducción Genética , Transgenes
5.
Stem Cells Transl Med ; 6(6): 1477-1490, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28225193

RESUMEN

Synucleinopathies are a group of neurodegenerative disorders sharing the common feature of misfolding and accumulation of the presynaptic protein α-synuclein (α-syn) into insoluble aggregates. Within this diverse group, Dementia with Lewy Bodies (DLB) is characterized by the aberrant accumulation of α-syn in cortical, hippocampal, and brainstem neurons, resulting in multiple cellular stressors that particularly impair dopamine and glutamate neurotransmission and related motor and cognitive function. Recent studies show that murine neural stem cell (NSC) transplantation can improve cognitive or motor function in transgenic models of Alzheimer's and Huntington's disease, and DLB. However, examination of clinically relevant human NSCs in these models is hindered by the challenges of xenotransplantation and the confounding effects of immunosuppressant drugs on pathology and behavior. To address this challenge, we developed an immune-deficient transgenic model of DLB that lacks T-, B-, and NK-cells, yet exhibits progressive accumulation of human α-syn (h-α-syn)-laden inclusions and cognitive and motor impairments. We demonstrate that clinically relevant human neural progenitor cells (line CNS10-hNPCs) survive, migrate extensively and begin to differentiate preferentially into astrocytes following striatal transplantation into this DLB model. Critically, grafted CNS10-hNPCs rescue both cognitive and motor deficits after 1 and 3 months and, furthermore, restore striatal dopamine and glutamate systems. These behavioral and neurochemical benefits are likely achieved by reducing α-syn oligomers. Collectively, these results using a new model of DLB demonstrate that hNPC transplantation can impact a broad array of disease mechanisms and phenotypes and suggest a cellular therapeutic strategy that should be pursued. Stem Cells Translational Medicine 2017;6:1477-1490.


Asunto(s)
Enfermedad por Cuerpos de Lewy/terapia , Células-Madre Neurales/trasplante , Trasplante de Células Madre/métodos , alfa-Sinucleína/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Células Cultivadas , Humanos , Memoria , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis
6.
Sci Rep ; 6: 25474, 2016 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-27146274

RESUMEN

In recent years, the assay for transposase-accessible chromatin using sequencing (ATAC-Seq) has become a fundamental tool of epigenomic research. However, it is difficult to perform this technique on frozen samples because freezing cells before extracting nuclei can impair nuclear integrity and alter chromatin structure, especially in fragile cells such as neurons. Our aim was to develop a protocol for freezing neuronal cells that is compatible with ATAC-Seq; we focused on a disease-relevant cell type, namely motor neurons differentiated from induced pluripotent stem cells (iMNs) from a patient affected by spinal muscular atrophy. We found that while flash-frozen iMNs are not suitable for ATAC-Seq, the assay is successful with slow-cooled cryopreserved cells. Using this method, we were able to isolate high quality, intact nuclei, and we verified that epigenetic results from fresh and cryopreserved iMNs quantitatively agree.


Asunto(s)
Núcleo Celular/efectos de los fármacos , Cromatina/efectos de los fármacos , Criopreservación/métodos , Crioprotectores/farmacología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Neuronas Motoras/efectos de los fármacos , Diferenciación Celular , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Supervivencia Celular/efectos de los fármacos , Cromatina/metabolismo , Cromatina/ultraestructura , Epigénesis Genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/patología , Cultivo Primario de Células , Análisis de Secuencia de ADN , Transposasas/química
7.
Exp Neurol ; 280: 41-9, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27032721

RESUMEN

Age-associated health decline presents a significant challenge to healthcare, although there are few animal models that can be used to test potential treatments. Here, we show that there is a significant reduction in both spinal cord motor neurons and motor function over time in the aging rat. One explanation for this motor neuron loss could be reduced support from surrounding aging astrocytes. Indeed, we have previously shown using in vitro models that aging rat astrocytes are less supportive to rat motor neuron function and survival over time. Here, we test whether rejuvenating the astrocyte niche can improve the survival of motor neurons in an aging spinal cord. We transplanted fetal-derived human neural progenitor cells (hNPCs) into the aging rat spinal cord and found that the cells survive and differentiate into astrocytes with a much higher efficiency than when transplanted into younger animals, suggesting that the aging environment stimulates astrocyte maturation. Importantly, the engrafted astrocytes were able to protect against motor neuron loss associated with aging, although this did not result in an increase in motor function based on behavioral assays. We also transplanted hNPCs genetically modified to secrete glial cell line-derived neurotrophic factor (GDNF) into the aging rat spinal cord, as this combination of cell and protein delivery can protect motor neurons in animal models of ALS. During aging, GDNF-expressing hNPCs protected motor neurons, though to the same extent as hNPCs alone, and again had no effect on motor function. We conclude that hNPCs can survive well in the aging spinal cord, protect motor neurons and mature faster into astrocytes when compared to transplantation into the young spinal cord. While there was no functional improvement, there were no functional deficits either, further supporting a good safety profile of hNPC transplantation even into the older patient population.


Asunto(s)
Envejecimiento/fisiología , Astrocitos/fisiología , Diferenciación Celular/fisiología , Neuronas Motoras/fisiología , Trastornos del Movimiento/cirugía , Células-Madre Neurales/fisiología , Factores de Edad , Animales , Peso Corporal/fisiología , Corteza Cerebral/citología , Modelos Animales de Enfermedad , Conducta Exploratoria/fisiología , Feto/citología , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Masculino , Trastornos del Movimiento/patología , Trastornos del Movimiento/fisiopatología , Fuerza Muscular/fisiología , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/trasplante , Unión Neuromuscular/fisiología , Ratas , Ratas Sprague-Dawley , Médula Espinal/citología , Médula Espinal/trasplante
8.
Transl Vis Sci Technol ; 4(1): 7, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25694843

RESUMEN

PURPOSE: Stem and progenitor cell transplantation provides a promising clinical application for treating degenerative retinal diseases, including age-related macular degeneration (AMD) and retinitis pigmentosa (RP). Our previous studies have shown that a single subretinal injection of human cortical-derived neural progenitor cells (hNPCctx) into cyclosporine-treated Royal College of Surgeons (RCS) rats preserved both photoreceptors and visual function. However, it is still unknown whether nonautologous progenitor cell readministration for sustained vision is efficacious and safe in terms of the initial graft initiating an immune response to a subsequent graft. METHODS: A cell suspension containing 3×104 hNPCctx into one eye of cyclosporine-treated RCS rats at postnatal day 21 (P21), followed by a second transplantation at P95 into the previously untreated fellow eye. RESULTS: hNPCctx delayed photoreceptor degeneration and preserved visual function, as measured by electroretinography (ERG), optokinetic response (OKR), and luminance threshold recordings (LTRs). Visual function and photoreceptors of the initially treated eye were still preserved 6 weeks after hNPCctx were injected into the second eye. Antibodies against T-cell markers showed that CD3, CD4, and CD8 T cells were not detected at P90 and P140 in most cases. No detectable level of anti-nestin antibody was found in serum by enzyme-linked immunosorbent assay (ELISA). CONCLUSIONS: This xenograft study with cyclosporine-treated animals demonstrates that readministration of hNPCctx into the fellow eye did not induce anti-graft immune responses or lower therapeutic efficacy of hNPCctx in preserving vision. Thus, readministration of progenitor cells to sustain long-term efficacy may be an option for long-term therapies of retinal degeneration. TRANSLATIONAL RELEVANCE: Redosing neural progenitors do not affect the efficacy of the initial grafts in protecting vision or induce unwanted immune responses.

9.
J Vis Exp ; (88)2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24962813

RESUMEN

A cell expansion technique to amass large numbers of cells from a single specimen for research experiments and clinical trials would greatly benefit the stem cell community. Many current expansion methods are laborious and costly, and those involving complete dissociation may cause several stem and progenitor cell types to undergo differentiation or early senescence. To overcome these problems, we have developed an automated mechanical passaging method referred to as "chopping" that is simple and inexpensive. This technique avoids chemical or enzymatic dissociation into single cells and instead allows for the large-scale expansion of suspended, spheroid cultures that maintain constant cell/cell contact. The chopping method has primarily been used for fetal brain-derived neural progenitor cells or neurospheres, and has recently been published for use with neural stem cells derived from embryonic and induced pluripotent stem cells. The procedure involves seeding neurospheres onto a tissue culture Petri dish and subsequently passing a sharp, sterile blade through the cells effectively automating the tedious process of manually mechanically dissociating each sphere. Suspending cells in culture provides a favorable surface area-to-volume ratio; as over 500,000 cells can be grown within a single neurosphere of less than 0.5 mm in diameter. In one T175 flask, over 50 million cells can grow in suspension cultures compared to only 15 million in adherent cultures. Importantly, the chopping procedure has been used under current good manufacturing practice (cGMP), permitting mass quantity production of clinical-grade cell products.


Asunto(s)
Encéfalo/citología , Células-Madre Neurales/citología , Células Madre Pluripotentes/citología , Encéfalo/embriología , Agregación Celular/fisiología , Comunicación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Técnicas Citológicas/métodos , Humanos
10.
Neuroreport ; 25(6): 367-72, 2014 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-24284956

RESUMEN

Human neural progenitor cells (hNPCs) derived from the fetal cortex can be expanded in vitro and genetically modified through lentiviral transduction to secrete growth factors shown to have a neurotrophic effect in animal models of neurological disease. hNPCs survive and mature following transplantation into the central nervous system of large and small animals including the rat model of amyotrophic lateral sclerosis. Here we report that hNPCs engineered to express glial cell line-derived neurotrophic factor (GDNF) survive long-term (7.5 months) following transplantation into the spinal cord of athymic nude rats and continue to secrete GDNF. Cell proliferation declined while the number of astrocytes increased, suggesting final maturation of the cells over time in vivo. Together these data show that GDNF-producing hNPCs may be useful as a source of cells for long-term delivery of both astrocytes and GDNF to the damaged central nervous system.


Asunto(s)
Células Madre Fetales/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Supervivencia de Injerto/fisiología , Células-Madre Neurales/metabolismo , Médula Espinal/trasplante , Animales , Diferenciación Celular/fisiología , Línea Celular , Supervivencia Celular/fisiología , Células Madre Fetales/trasplante , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Masculino , Células-Madre Neurales/trasplante , Ratas , Ratas Desnudas , Médula Espinal/patología
11.
Stem Cell Res ; 10(3): 417-427, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23474892

RESUMEN

We have developed a simple method to generate and expand multipotent, self-renewing pre-rosette neural stem cells from both human embryonic stem cells (hESCs) and human induced pluripotent stem cells (iPSCs) without utilizing embryoid body formation, manual selection techniques, or complex combinations of small molecules. Human ESC and iPSC colonies were lifted and placed in a neural stem cell medium containing high concentrations of EGF and FGF-2. Cell aggregates (termed EZ spheres) could be expanded for long periods using a chopping method that maintained cell-cell contact. Early passage EZ spheres rapidly down-regulated OCT4 and up-regulated SOX2 and nestin expression. They retained the potential to form neural rosettes and consistently differentiated into a range of central and peripheral neural lineages. Thus, they represent a very early neural stem cell with greater differentiation flexibility than other previously described methods. As such, they will be useful for the rapidly expanding field of neurological development and disease modeling, high-content screening, and regenerative therapies based on pluripotent stem cell technology.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células Madre Multipotentes/citología , Células-Madre Neurales/citología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo/química , Factor de Crecimiento Epidérmico/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Humanos , Proteínas de Filamentos Intermediarios/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Nestina , Células-Madre Neurales/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Factores de Transcripción SOXB1/metabolismo , Regulación hacia Arriba
12.
PLoS One ; 4(10): e7630, 2009 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-19898616

RESUMEN

BACKGROUND: Stem cell expansion and differentiation is the foundation of emerging cell therapy technologies. The potential applications of human neural progenitor cells (hNPCs) are wide ranging, but a normal cytogenetic profile is important to avoid the risk of tumor formation in clinical trials. FDA approved clinical trials are being planned and conducted for hNPC transplantation into the brain or spinal cord for various neurodegenerative disorders. Although human embryonic stem cells (hESCs) are known to show recurrent chromosomal abnormalities involving 12 and 17, no studies have revealed chromosomal abnormalities in cultured hNPCs. Therefore, we investigated frequently occurring chromosomal abnormalities in 21 independent fetal-derived hNPC lines and the possible mechanisms triggering such aberrations. METHODS AND FINDINGS: While most hNPC lines were karyotypically normal, G-band karyotyping and fluorescent in situ hybridization (FISH) analyses revealed the emergence of trisomy 7 (hNPC(+7)) and trisomy 19 (hNPC(+19)), in 24% and 5% of the lines, respectively. Once detected, subsequent passaging revealed emerging dominance of trisomy hNPCs. DNA microarray and immunoblotting analyses demonstrate epidermal growth factor receptor (EGFR) overexpression in hNPC(+7) and hNPC(+19) cells. We observed greater levels of telomerase (hTERT), increased proliferation (Ki67), survival (TUNEL), and neurogenesis (beta(III)-tubulin) in hNPC(+7) and hNPC(+19), using respective immunocytochemical markers. However, the trisomy lines underwent replicative senescence after 50-60 population doublings and never showed neoplastic changes. Although hNPC(+7) and hNPC(+19) survived better after xenotransplantation into the rat striatum, they did not form malignant tumors. Finally, EGF deprivation triggered a selection of trisomy 7 cells in a diploid hNPC line. CONCLUSIONS: We report that hNPCs are susceptible to accumulation of chromosome 7 and 19 trisomy in long-term cell culture. These results suggest that micro-environmental cues are powerful factors in the selection of specific hNPC aneuploidies, with trisomy of chromosome 7 being the most common. Given that a number of stem cell based clinical trials are being conducted or planned in USA and a recent report in PLoS Medicine showing the dangers of grafting an inordinate number of cells, these data substantiate the need for careful cytogenetic evaluation of hNPCs (fetal or hESC-derived) before their use in clinical or basic science applications.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Cromosomas Humanos Par 19/ultraestructura , Cromosomas Humanos Par 7/ultraestructura , Neuronas/citología , Células Madre/citología , Trisomía , Encéfalo/embriología , Aberraciones Cromosómicas , Citogenética , Células Madre Embrionarias/citología , Receptores ErbB/metabolismo , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Análisis de Secuencia por Matrices de Oligonucleótidos
13.
Mol Ther ; 16(12): 2002-10, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18797452

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease in which there is a progressive loss of motor neurons and their connections to muscle, leading to paralysis. In order to maintain muscle connections in a rat model of familial ALS (FALS), we performed intramuscular transplantation with human mesenchymal stem cells (hMSCs) used as "Trojan horses" to deliver growth factors to the terminals of motor neurons and to the skeletal muscles. hMSCs engineered to secrete glial cell line-derived neurotrophic factor (hMSC-GDNF) were transplanted bilaterally into three muscle groups. The cells survived within the muscle, released GDNF, and significantly increased the number of neuromuscular connections and motor neuron cell bodies in the spinal cord at mid-stages of the disease. Further, intramuscular transplantation with hMSC-GDNF was found to ameliorate motor neuron loss within the spinal cord where it connects with the limb muscles receiving transplants. While disease onset was similar in all the animals, hMSC-GDNF significantly delayed disease progression, increasing overall lifespan by up to 28 days, which is one of the largest effects on survival noted for this rat model of FALS. This preclinical data provides a novel and practical approach toward ex vivo gene therapy for ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Células Madre Mesenquimatosas/metabolismo , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Músculos/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/terapia , Animales , Supervivencia Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Terapia Genética , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Humanos , Trasplante de Células Madre Mesenquimatosas , Mutación/genética , Neuroglía/metabolismo , Unión Neuromuscular/genética , Unión Neuromuscular/metabolismo , Ratas , Ratas Sprague-Dawley , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1 , Tasa de Supervivencia
14.
PLoS One ; 2(8): e689, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17668067

RESUMEN

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a fatal, progressive neurodegenerative disease characterized by rapid loss of muscle control and eventual paralysis due to the death of large motor neurons in the brain and spinal cord. Growth factors such as glial cell line derived neurotrophic factor (GDNF) are known to protect motor neurons from damage in a range of models. However, penetrance through the blood brain barrier and delivery to the spinal cord remains a serious challenge. Although there may be a primary dysfunction in the motor neuron itself, there is also increasing evidence that excitotoxicity due to glial dysfunction plays a crucial role in disease progression. Clearly it would be of great interest if wild type glial cells could ameliorate motor neuron loss in these models, perhaps in combination with the release of growth factors such as GDNF. METHODOLOGY/PRINCIPAL FINDINGS: Human neural progenitor cells can be expanded in culture for long periods and survive transplantation into the adult rodent central nervous system, in some cases making large numbers of GFAP positive astrocytes. They can also be genetically modified to release GDNF (hNPC(GDNF)) and thus act as long-term 'mini pumps' in specific regions of the rodent and primate brain. In the current study we genetically modified human neural stem cells to release GDNF and transplanted them into the spinal cord of rats over-expressing mutant SOD1 (SOD1(G93A)). Following unilateral transplantation into the spinal cord of SOD1(G93A) rats there was robust cellular migration into degenerating areas, efficient delivery of GDNF and remarkable preservation of motor neurons at early and end stages of the disease within chimeric regions. The progenitors retained immature markers, and those not secreting GDNF had no effect on motor neuron survival. Interestingly, this robust motor neuron survival was not accompanied by continued innervation of muscle end plates and thus resulted in no improvement in ipsilateral limb use. CONCLUSIONS/SIGNIFICANCE: The potential to maintain dying motor neurons by delivering GDNF using neural progenitor cells represents a novel and powerful treatment strategy for ALS. While this approach represents a unique way to prevent motor neuron loss, our data also suggest that additional strategies may also be required for maintenance of neuromuscular connections and full functional recovery. However, simply maintaining motor neurons in patients would be the first step of a therapeutic advance for this devastating and incurable disease, while future strategies focus on the maintenance of the neuromuscular junction.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Neuronas Motoras/metabolismo , Músculos/inervación , Células Madre/fisiología , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Astrocitos/citología , Astrocitos/metabolismo , Conducta Animal/fisiología , Biomarcadores/metabolismo , Movimiento Celular , Supervivencia Celular/fisiología , Células Cultivadas , Femenino , Feto/citología , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Actividad Motora/fisiología , Neuronas Motoras/citología , Neuronas Motoras/patología , Unión Neuromuscular/ultraestructura , Ratas , Ratas Sprague-Dawley , Médula Espinal/citología , Médula Espinal/patología , Trasplante de Células Madre , Células Madre/citología , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1
15.
Amyotroph Lateral Scler ; 8(1): 20-5, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17364431

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease causing the progressive loss of brain and spinal cord motor neurons. The exact etiology of ALS is still uncertain, but males have consistently been shown to be at a higher risk for the disease than females. Recently, transgenic rats overexpressing mutant forms of the human SOD1 (hSOD1) gene have been established as a valuable disease model of ALS. Here we show that sexual dimorphism in disease onset is also observed in hSOD1G93A transgenic rats. Disease onset was consistently earlier in male than in female hSOD1G93A rats. We also found that hSOD1G93A male rats lost weight more rapidly following disease onset compared to hSOD1G93A females. Furthermore, we tested locomotor function using the Basso-Beattie-Bresnahan (BBB) rating scale and a beam walking test. We found that motor dysfunction started earlier in males than in females but progressed similarly in the two sexes. These results have important implications for future experimentation and therapeutic development using the rat model of ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/etiología , Esclerosis Amiotrófica Lateral/fisiopatología , Modelos Animales de Enfermedad , Caracteres Sexuales , Factores de Edad , Edad de Inicio , Esclerosis Amiotrófica Lateral/genética , Análisis de Varianza , Animales , Animales Modificados Genéticamente , Conducta Animal , Peso Corporal , Progresión de la Enfermedad , Actividad Motora/fisiología , Desempeño Psicomotor/fisiología , Ratas , Ratas Sprague-Dawley , Factores Sexuales , Superóxido Dismutasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA