Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 40(10): e106389, 2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-33792056

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder whose exact causative mechanisms are still under intense investigation. Several lines of evidence suggest that the anatomical and temporal propagation of pathological protein species along the neural axis could be among the main driving mechanisms for the fast and irreversible progression of ALS pathology. Many ALS-associated proteins form intracellular aggregates as a result of their intrinsic prion-like properties and/or following impairment of the protein quality control systems. During the disease course, these mutated proteins and aberrant peptides are released in the extracellular milieu as soluble or aggregated forms through a variety of mechanisms. Internalization by recipient cells may seed further aggregation and amplify existing proteostatic imbalances, thus triggering a vicious cycle that propagates pathology in vulnerable cells, such as motor neurons and other susceptible neuronal subtypes. Here, we provide an in-depth review of ALS pathology with a particular focus on the disease mechanisms of seeding and transmission of the most common ALS-associated proteins, including SOD1, FUS, TDP-43, and C9orf72-linked dipeptide repeats. For each of these proteins, we report historical, biochemical, and pathological evidence of their behaviors in ALS. We further discuss the possibility to harness pathological proteins as biomarkers and reflect on the implications of these findings for future research.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Neuronas Motoras/metabolismo , Esclerosis Amiotrófica Lateral/genética , Proteínas de Unión al ADN/metabolismo , Endocitosis/fisiología , Exocitosis/fisiología , Humanos , Pliegue de Proteína , Proteína FUS de Unión a ARN/metabolismo , Superóxido Dismutasa-1/metabolismo
2.
Hum Mol Genet ; 31(16): 2693-2710, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-35313342

RESUMEN

Hereditary spastic paraplegia type 15 (HSP15) is a neurodegenerative condition caused by the inability to produce SPG15 protein, which leads to lysosomal swelling. However, the link between lysosomal aberrations and neuronal death is poorly explored. To uncover the functional consequences of lysosomal aberrations in disease pathogenesis, we analyze human dermal fibroblasts from HSP15 patients as well as primary cortical neurons derived from an SPG15 knockout (KO) mouse model. We find that SPG15 protein loss induces defective anterograde transport, impaired neurite outgrowth, axonal swelling and reduced autophagic flux in association with the onset of lysosomal abnormalities. Additionally, we observe lipid accumulation within the lysosomal compartment, suggesting that distortions in cellular lipid homeostasis are intertwined with lysosomal alterations. We further demonstrate that SPG15 KO neurons exhibit synaptic dysfunction, accompanied by augmented vulnerability to glutamate-induced excitotoxicity. Overall, our study establishes an intimate link between lysosomal aberrations, lipid metabolism and electrophysiological impairments, suggesting that lysosomal defects are at the core of multiple neurodegenerative disease processes in HSP15.


Asunto(s)
Enfermedades Neurodegenerativas , Paraplejía Espástica Hereditaria , Animales , Proteínas Portadoras/metabolismo , Humanos , Metabolismo de los Lípidos/genética , Lípidos , Lisosomas/metabolismo , Ratones , Enfermedades Neurodegenerativas/metabolismo , Proteínas/metabolismo , Degeneración Retiniana , Paraplejía Espástica Hereditaria/metabolismo
3.
Acta Neuropathol ; 138(1): 67-84, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30937520

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a lethal disease characterized by motor neuron degeneration and associated with aggregation of nuclear RNA-binding proteins (RBPs), including FUS. How FUS aggregation and neurodegeneration are prevented in healthy motor neurons remain critically unanswered questions. Here, we use a combination of ALS patient autopsy tissue and induced pluripotent stem cell-derived neurons to study the effects of FUS mutations on RBP homeostasis. We show that FUS' tendency to aggregate is normally buffered by interacting RBPs, but this buffering is lost when FUS mislocalizes to the cytoplasm due to ALS mutations. The presence of aggregation-prone FUS in the cytoplasm causes imbalances in RBP homeostasis that exacerbate neurodegeneration. However, enhancing autophagy using small molecules reduces cytoplasmic FUS, restores RBP homeostasis and rescues motor function in vivo. We conclude that disruption of RBP homeostasis plays a critical role in FUS-ALS and can be treated by stimulating autophagy.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Autofagia/fisiología , Neuronas Motoras/patología , Citoplasma/metabolismo , Humanos , Cuerpos de Inclusión/patología , Células Madre Pluripotentes Inducidas/patología , Mutación/genética , Proteína FUS de Unión a ARN/metabolismo
4.
Development ; 142(17): 2904-15, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26329599

RESUMEN

The mammalian neocortex shows a conserved six-layered structure that differs between species in the total number of cortical neurons produced owing to differences in the relative abundance of distinct progenitor populations. Recent studies have identified a new class of proliferative neurogenic cells in the outer subventricular zone (OSVZ) in gyrencephalic species such as primates and ferrets. Lissencephalic brains of mice possess fewer OSVZ-like progenitor cells and these do not constitute a distinct layer. Most in vitro and in vivo studies have shown that oxygen regulates the maintenance, proliferation and differentiation of neural progenitor cells. Here we dissect the effects of fetal brain oxygen tension on neural progenitor cell activity using a novel mouse model that allows oxygen tension to be controlled within the hypoxic microenvironment in the neurogenic niche of the fetal brain in vivo. Indeed, maternal oxygen treatment of 10%, 21% and 75% atmospheric oxygen tension for 48 h translates into robust changes in fetal brain oxygenation. Increased oxygen tension in fetal mouse forebrain in vivo leads to a marked expansion of a distinct proliferative cell population, basal to the SVZ. These cells constitute a novel neurogenic cell layer, similar to the OSVZ, and contribute to corticogenesis by heading for deeper cortical layers as a part of the cortical plate.


Asunto(s)
Ventrículos Laterales/embriología , Ventrículos Laterales/patología , Oxígeno/farmacología , Células Madre/patología , Animales , Recuento de Células , Proliferación Celular/efectos de los fármacos , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Feto/efectos de los fármacos , Feto/metabolismo , Hiperoxia/embriología , Hiperoxia/patología , Ventrículos Laterales/irrigación sanguínea , Ventrículos Laterales/efectos de los fármacos , Ratones Endogámicos C57BL , Mitosis/efectos de los fármacos , Modelos Biológicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Tamaño de los Órganos/efectos de los fármacos , Prosencéfalo/efectos de los fármacos , Prosencéfalo/embriología , Prosencéfalo/metabolismo , Prosencéfalo/patología , Factores de Transcripción SOXB1/metabolismo , Células Madre/efectos de los fármacos , Proteínas de Dominio T Box/metabolismo
5.
Expert Opin Biol Ther ; 22(9): 1163-1176, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-34904932

RESUMEN

INTRODUCTION: Gene therapy provides the exciting opportunity of a curative single treatment for devastating diseases, eradicating the need for chronic medication. Adeno-associated viruses (AAVs) are among the most attractive vector carriers for gene replacement in vivo. Yet, despite the success of recent AAV-based clinical trials, the clinical use of these vectors has been limited. For instance, the AAV packaging capacity is restricted to ~4.7 kb, making it a substantial challenge to deliver large gene products. AREAS COVERED: In this review, we explore established and emerging strategies that circumvent the packaging limit of AAVs to make them effective vehicles for gene replacement therapy of monogenic disorders, with a particular focus on diseases affecting the nervous system. We report historical references, design remarks, as well as strengths and weaknesses of these approaches. We additionally discuss examples of neurological disorders for which such strategies have been attempted. EXPERT OPINION: The field of AAV-gene therapy has experienced enormous advancements in the last decade. However, there is still ample space for improvement aimed at overcoming existing challenges that are slowing down the progressive trajectory of this field.


Asunto(s)
Vectores Genéticos , Enfermedades del Sistema Nervioso , Dependovirus/genética , Terapia Genética , Humanos , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/terapia
6.
Life Sci Alliance ; 5(8)2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35440492

RESUMEN

Spinal muscular atrophy, the leading genetic cause of infant mortality, is a motor neuron disease caused by low levels of survival motor neuron (SMN) protein. SMN is a multifunctional protein that is implicated in numerous cytoplasmic and nuclear processes. Recently, increasing attention is being paid to the role of SMN in the maintenance of DNA integrity. DNA damage and genome instability have been linked to a range of neurodegenerative diseases. The ribosomal DNA (rDNA) represents a particularly unstable locus undergoing frequent breakage. Instability in rDNA has been associated with cancer, premature ageing syndromes, and a number of neurodegenerative disorders. Here, we report that SMN-deficient cells exhibit increased rDNA damage leading to impaired ribosomal RNA synthesis and translation. We also unravel an interaction between SMN and RNA polymerase I. Moreover, we uncover an spinal muscular atrophy motor neuron-specific deficiency of DDX21 protein, which is required for resolving R-loops in the nucleolus. Taken together, our findings suggest a new role of SMN in rDNA integrity.


Asunto(s)
Neuronas Motoras , Atrofia Muscular Espinal , ARN Helicasas DEAD-box/metabolismo , Daño del ADN/genética , ADN Ribosómico/genética , ADN Ribosómico/metabolismo , Humanos , Lactante , Neuronas Motoras/metabolismo , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Ribosomas/genética , Ribosomas/metabolismo
7.
Life Sci Alliance ; 5(9)2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35568435

RESUMEN

Dipeptide repeat (DPR) proteins are aggregation-prone polypeptides encoded by the pathogenic GGGGCC repeat expansion in the C9ORF72 gene, the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia. In this study, we focus on the role of poly-GA DPRs in disease spread. We demonstrate that recombinant poly-GA oligomers can directly convert into solid-like aggregates and form characteristic ß-sheet fibrils in vitro. To dissect the process of cell-to-cell DPR transmission, we closely follow the fate of poly-GA DPRs in either their oligomeric or fibrillized form after administration in the cell culture medium. We observe that poly-GA DPRs are taken up via dynamin-dependent and -independent endocytosis, eventually converging at the lysosomal compartment and leading to axonal swellings in neurons. We then use a co-culture system to demonstrate astrocyte-to-motor neuron DPR propagation, showing that astrocytes may internalise and release aberrant peptides in disease pathogenesis. Overall, our results shed light on the mechanisms of poly-GA cellular uptake and propagation, suggesting lysosomal impairment as a possible feature underlying the cellular pathogenicity of these DPR species.


Asunto(s)
Esclerosis Amiotrófica Lateral , Proteína C9orf72 , Demencia Frontotemporal , Esclerosis Amiotrófica Lateral/patología , Proteína C9orf72/genética , Dipéptidos , Demencia Frontotemporal/genética , Demencia Frontotemporal/metabolismo , Demencia Frontotemporal/patología , Humanos , Neuronas Motoras/metabolismo
8.
Methods Mol Biol ; 1888: 21-43, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30519939

RESUMEN

High-throughput phenotypic screening enables the identification of new therapeutic targets even when the molecular mechanism underlying the disease is unknown. In the case of neurodegenerative disease, there is a dire need to identify new targets that can ameliorate, halt, or reverse degeneration. Stem cell-based disease models are particularly powerful tools for phenotypic screening because they use the same cell type affected in patients. Here, we describe the expansion of mouse stem cells and human induced pluripotent stem cells as well as the differentiation of these cells into neural lineages that, when exposed to neuroinflammatory stress, can be used for compound screening followed by hit identification, validation, and target deconvolution.


Asunto(s)
Técnicas de Cultivo de Célula , Descubrimiento de Drogas , Fenotipo , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Diferenciación Celular , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Reproducibilidad de los Resultados , Bibliotecas de Moléculas Pequeñas , Células Madre/citología
9.
Nat Commun ; 10(1): 5583, 2019 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-31811140

RESUMEN

Mutations in fused in sarcoma (FUS) lead to amyotrophic lateral sclerosis (ALS) with varying ages of onset, progression and severity. This suggests that unknown genetic factors contribute to disease pathogenesis. Here we show the identification of muscleblind as a novel modifier of FUS-mediated neurodegeneration in vivo. Muscleblind regulates cytoplasmic mislocalization of mutant FUS and subsequent accumulation in stress granules, dendritic morphology and toxicity in mammalian neuronal and human iPSC-derived neurons. Interestingly, genetic modulation of endogenous muscleblind was sufficient to restore survival motor neuron (SMN) protein localization in neurons expressing pathogenic mutations in FUS, suggesting a potential mode of suppression of FUS toxicity. Upregulation of SMN suppressed FUS toxicity in Drosophila and primary cortical neurons, indicating a link between FUS and SMN. Our data provide in vivo evidence that muscleblind is a dominant modifier of FUS-mediated neurodegeneration by regulating FUS-mediated ALS pathogenesis.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas Nucleares/metabolismo , Proteína FUS de Unión a ARN/metabolismo , Proteínas del Complejo SMN/metabolismo , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Esclerosis Amiotrófica Lateral/genética , Animales , Citoplasma/metabolismo , Gránulos Citoplasmáticos/metabolismo , Drosophila/genética , Drosophila/metabolismo , Femenino , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Neuronas Motoras/metabolismo , Mutación , Fenotipo , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/toxicidad , Proteínas del Complejo SMN/genética , Factores de Transcripción/metabolismo
10.
Stem Cell Reports ; 12(3): 502-517, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30773488

RESUMEN

Neuroinflammation is a hallmark of neurological disorders and is accompanied by the production of neurotoxic agents such as nitric oxide. We used stem cell-based phenotypic screening and identified small molecules that directly protected neurons from neuroinflammation-induced degeneration. We demonstrate that inhibition of CDK5 is involved in, but not sufficient for, neuroprotection. Instead, additional inhibition of GSK3ß is required to enhance the neuroprotective effects of CDK5 inhibition, which was confirmed using short hairpin RNA-mediated knockdown of CDK5 and GSK3ß. Quantitative phosphoproteomics and high-content imaging demonstrate that neurite degeneration is mediated by aberrant phosphorylation of multiple microtubule-associated proteins. Finally, we show that our hit compound protects neurons in vivo in zebrafish models of motor neuron degeneration and Alzheimer's disease. Thus, we demonstrate an overlap of CDK5 and GSK3ß in mediating the regulation of the neuronal cytoskeleton and that our hit compound LDC8 represents a promising starting point for neuroprotective drugs.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , Citoesqueleto/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Inflamación/metabolismo , Degeneración Nerviosa/metabolismo , Neuronas/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Animales , Citoesqueleto/efectos de los fármacos , Humanos , Inflamación/tratamiento farmacológico , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Degeneración Nerviosa/tratamiento farmacológico , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Pez Cebra/metabolismo
11.
PLoS One ; 13(3): e0192497, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29513666

RESUMEN

Induced pluripotent stem cells (iPSCs) have recapitulated several aspects of Parkinson's disease (PD), but most iPSCs are derived from familial cases, which account for only about 15% of patients. Thus, while the emphasis has justifiably been on using iPSCs to model rare familial cases, models for the most common forms of PD are critically lacking. Here, we report the generation of an iPSC-based model of idiopathic PD (iPD) with or without RS1491923, which is a common risk variant in the LRRK2 locus. Consistent with GWA studies, we found large variability in our datasets. However, iPSC-derived neurons carrying the risk allele emerged for displaying subtle disturbances of cellular degradative systems, in line with familial PD models. We also observed that treatment with the LRRK2 inhibitor CZC-25146 slightly reduced a marker of aSYN pathology in all iPD lines. Future iPSC-based studies may need to be structured similarly to large GWA studies in order to obtain relevant statistical power. However, results from this pilot study suggest that iPSC-based modeling represents an attractive way to investigate idiopathic diseases.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Enfermedad de Parkinson/genética , Polimorfismo de Nucleótido Simple , Anciano , Alelos , Diferenciación Celular/genética , Células Cultivadas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Modelos Genéticos , Enfermedad de Parkinson/patología , Proyectos Piloto , Factores de Riesgo
12.
Science ; 360(6391): 918-921, 2018 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-29650702

RESUMEN

Prion-like RNA binding proteins (RBPs) such as TDP43 and FUS are largely soluble in the nucleus but form solid pathological aggregates when mislocalized to the cytoplasm. What keeps these proteins soluble in the nucleus and promotes aggregation in the cytoplasm is still unknown. We report here that RNA critically regulates the phase behavior of prion-like RBPs. Low RNA/protein ratios promote phase separation into liquid droplets, whereas high ratios prevent droplet formation in vitro. Reduction of nuclear RNA levels or genetic ablation of RNA binding causes excessive phase separation and the formation of cytotoxic solid-like assemblies in cells. We propose that the nucleus is a buffered system in which high RNA concentrations keep RBPs soluble. Changes in RNA levels or RNA binding abilities of RBPs cause aberrant phase transitions.


Asunto(s)
Núcleo Celular/química , Citoplasma/química , Priones/química , Agregación Patológica de Proteínas/metabolismo , ARN Nuclear/química , Proteínas de Unión al ARN/química , Células HeLa , Humanos , Gotas Lipídicas , Transición de Fase , Agregado de Proteínas , Solubilidad
13.
Stem Cell Reports ; 10(2): 375-389, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29358088

RESUMEN

Perturbations in stress granule (SG) dynamics may be at the core of amyotrophic lateral sclerosis (ALS). Since SGs are membraneless compartments, modeling their dynamics in human motor neurons has been challenging, thus hindering the identification of effective therapeutics. Here, we report the generation of isogenic induced pluripotent stem cells carrying wild-type and P525L FUS-eGFP. We demonstrate that FUS-eGFP is recruited into SGs and that P525L profoundly alters their dynamics. With a screening campaign, we demonstrate that PI3K/AKT/mTOR pathway inhibition increases autophagy and ameliorates SG phenotypes linked to P525L FUS by reducing FUS-eGFP recruitment into SGs. Using a Drosophila model of FUS-ALS, we corroborate that induction of autophagy significantly increases survival. Finally, by screening clinically approved drugs for their ability to ameliorate FUS SG phenotypes, we identify a number of brain-penetrant anti-depressants and anti-psychotics that also induce autophagy. These drugs could be repurposed as potential ALS treatments.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Proteínas de Drosophila/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo F-H/genética , Células Madre Pluripotentes Inducidas/metabolismo , Proteína FUS de Unión a ARN/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Antidepresivos/farmacología , Antipiréticos/farmacología , Autofagia/genética , Sistemas CRISPR-Cas , Drosophila , Evaluación Preclínica de Medicamentos , Proteínas Fluorescentes Verdes/genética , Humanos , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Mutación , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/genética
15.
Front Cell Neurosci ; 11: 284, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28959189

RESUMEN

Endogenous oligodendrocyte progenitor cells (OPCs) are a promising target to improve functional recovery after spinal cord injury (SCI) by remyelinating denuded, and therefore vulnerable, axons. Demyelination is the result of a primary insult and secondary injury, leading to conduction blocks and long-term degeneration of the axons, which subsequently can lead to the loss of their neurons. In response to SCI, dormant OPCs can be activated and subsequently start to proliferate and differentiate into mature myelinating oligodendrocytes (OLs). Therefore, researchers strive to control OPC responses, and utilize small molecule screening approaches in order to identify mechanisms of OPC activation, proliferation, migration and differentiation. In zebrafish, OPCs remyelinate axons of the optic tract after lysophosphatidylcholine (LPC)-induced demyelination back to full thickness myelin sheaths. In contrast to zebrafish, mammalian OPCs are highly vulnerable to excitotoxic stress, a cause of secondary injury, and remyelination remains insufficient. Generally, injury induced remyelination leads to shorter internodes and thinner myelin sheaths in mammals. In this study, we show that myelin sheaths are lost early after a complete spinal transection injury, but are re-established within 14 days after lesion. We introduce a novel, easy-to-use, inexpensive and highly reproducible OPC culture system based on dormant spinal OPCs from adult zebrafish that enables in vitro analysis. Zebrafish OPCs are robust, can easily be purified with high viability and taken into cell culture. This method enables to examine why zebrafish OPCs remyelinate better than their mammalian counterparts, identify cell intrinsic responses, which could lead to pro-proliferating or pro-differentiating strategies, and to test small molecule approaches. In this methodology paper, we show efficient isolation of OPCs from adult zebrafish spinal cord and describe culture conditions that enable analysis up to 10 days in vitro. Finally, we demonstrate that zebrafish OPCs differentiate into Myelin Basic Protein (MBP)-expressing OLs when co-cultured with human motor neurons differentiated from induced pluripotent stem cells (iPSCs). This shows that the basic mechanisms of oligodendrocyte differentiation are conserved across species and that understanding the regulation of zebrafish OPCs can contribute to the development of new treatments to human diseases.

16.
Stem Cells Dev ; 25(3): 227-38, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26577812

RESUMEN

Oxygen tension is an important factor controlling stem cell proliferation and maintenance in various stem cell populations with a particular relevance in midbrain dopaminergic progenitors. Further studies have shown that the oxygen-dependent transcription factor hypoxia-inducible factor 1α (HIF-1α) is involved in these processes. However, all available studies on oxygen effects in dopaminergic neuroprogenitors were performed in vitro and thus it remains unclear whether tissue oxygen tension in the embryonic midbrain is also relevant for the regulation of dopaminergic neurogenesis in vivo. We thus dissect here the effects of oxygen tension in combination with HIF-1α conditional knockout on dopaminergic neurogenesis by using a novel experimental design allowing for the control of oxygen tension within the microenvironment of the neurogenic niche of the murine fetal midbrain in vivo. The microenvironment of the midbrain dopaminergic neurogenic niche was detected as hypoxic with oxygen tensions below 1.1%. Maternal oxygen treatment of 10%, 21%, and 75% atmospheric oxygen tension for 48 h translates into robust changes in fetal midbrain oxygenation. Fetal midbrain hypoxia hampered the generation of dopaminergic neurons and is accompanied with restricted fetal midbrain development. In contrast, induced hyperoxia stimulated proliferation and differentiation of dopaminergic progenitors during early and late embryogenesis. Oxygen effects were not directly mediated through HIF-1α signaling. These data--in agreement with in vitro data-indicate that oxygen is a crucial regulator of developmental dopaminergic neurogenesis. Our study provides the initial framework for future studies on molecular mechanisms mediating oxygen regulation of dopaminergic neurogenesis within the fetal midbrain as its natural environment.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Hipoxia Fetal/metabolismo , Mesencéfalo/metabolismo , Neurogénesis , Oxígeno/metabolismo , Nicho de Células Madre , Animales , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/patología , Hipoxia Fetal/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mesencéfalo/citología , Mesencéfalo/embriología , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología
17.
Future Microbiol ; 10(4): 665-82, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25865200

RESUMEN

Viral infections and opportunistic fungal pathogens represent a major menace for immunocompromised patients. Despite the availability of antifungal and antiviral drugs, mortality in these patients remains high, underlining the need of novel therapeutic options based on completely different strategies. This review describes the potential of several T-cell-based therapeutic approaches in the prophylaxis and treatment of infectious diseases with a particular focus on persistent viral infections and opportunistic fungal infections, as these mostly affect immunocompromised patients.


Asunto(s)
Traslado Adoptivo , Micosis/terapia , Infecciones Oportunistas/terapia , Linfocitos T/inmunología , Virosis/terapia , Humanos , Huésped Inmunocomprometido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA