Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Contemp Clin Trials ; 134: 107333, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37739167

RESUMEN

INTRODUCTION: Multiple sclerosis (MS) is a major cause of disability in young and middle-aged people, and myelin repair therapies are needed to slow or potentially reverse this damage. Bazedoxifene (BZA) is a selective estrogen receptor modulator identified in a novel high-throughput unbiased screen for its remyelinating potential, and its remyelinating effects were demonstrated in pre-clinical models. METHODS: This is a single-center, double blind, randomized, controlled, delayed-start Phase 2 clinical trial (NCT04002934) investigating the remyelinating effects of BZA relative to placebo. Female patients with relapsing-remitting MS, aged 45-60 years (or > 40 if post-menopausal), and ambulatory status (EDSS 0-6 inclusive), will be recruited into a clinical trial with 2 arms of identical design, except that the "Chronic Optic Neuropathy" arm requires additional inclusion criteria of electrophysiological evidence of prior visual pathway demyelination. Clinical, electrophysiological, and imaging evaluations will occur at baseline, 3 months, and 6 months. The primary outcome is change in Myelin Water Fraction (MWF) on MRI within the corpus callosum. Secondary outcomes are: visual evoked potential (VEP) P100 latency, novel digital measures of cognition and activity, and patient reported outcomes. Tertiary outcomes are: safety and tolerability. DISCUSSION: BZA has strong preclinical effects on myelin repair, and in the general population demonstrated benefits in treating postmenopausal osteoporosis. Together, these findings support the rationale for an RCT testing BZA in women with MS, evaluating established neuroimaging and neurovisual measures of myelin repair. Additionally, validating novel digital tools could increase sensitivity to change and inform the duration and design of future clinical trials.


Asunto(s)
Esclerosis Múltiple , Remielinización , Persona de Mediana Edad , Humanos , Femenino , Vaina de Mielina , Potenciales Evocados Visuales
2.
Nat Neurosci ; 23(4): 487-499, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32042175

RESUMEN

Experience-dependent myelination is hypothesized to shape neural circuit function and subsequent behavioral output. Using a contextual fear memory task in mice, we demonstrate that fear learning induces oligodendrocyte precursor cells to proliferate and differentiate into myelinating oligodendrocytes in the medial prefrontal cortex. Transgenic animals that cannot form new myelin exhibit deficient remote, but not recent, fear memory recall. Recording population calcium dynamics by fiber photometry, we observe that the neuronal response to conditioned context cues evolves over time in the medial prefrontal cortex, but not in animals that cannot form new myelin. Finally, we demonstrate that pharmacological induction of new myelin formation with clemastine fumarate improves remote memory recall and promotes fear generalization. Thus, bidirectional manipulation of myelin plasticity functionally affects behavior and neurophysiology, which suggests that neural activity during fear learning instructs the formation of new myelin, which in turn supports the consolidation and/or retrieval of remote fear memories.


Asunto(s)
Proliferación Celular/fisiología , Condicionamiento Clásico/fisiología , Miedo/fisiología , Memoria a Largo Plazo/fisiología , Vaina de Mielina/fisiología , Células Precursoras de Oligodendrocitos/fisiología , Animales , Ratones , Ratones Transgénicos , Factor de Transcripción 2 de los Oligodendrocitos/genética , Corteza Prefrontal/fisiología
3.
J Neurosci ; 39(12): 2184-2194, 2019 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-30696729

RESUMEN

A significant unmet need for patients with multiple sclerosis (MS) is the lack of U.S. Food and Drug Administration (FDA)-approved remyelinating therapies. We have identified a compelling remyelinating agent, bazedoxifene (BZA), a European Medicines Agency (EMA)-approved (and FDA-approved in combination with conjugated estrogens) selective estrogen receptor (ER) modulator (SERM) that could move quickly from bench to bedside. This therapy stands out as a tolerable alternative to previously identified remyelinating agents and other candidates within this family. Using an unbiased high-throughput screen, with subsequent validation in both murine and human oligodendrocyte precursor cells (OPCs) and coculture systems, we find that BZA enhances differentiation of OPCs into functional oligodendrocytes. Using an in vivo murine model of focal demyelination, we find that BZA enhances OPC differentiation and remyelination. Of critical importance, we find that BZA acts independently of its presumed target, the ER, in both in vitro and in vivo systems. Using a massive computational data integration approach, we independently identify six possible candidate targets through which SERMs may mediate their effect on remyelination. Of particular interest, we identify EBP (encoding 3ß-hydroxysteroid-Δ8,Δ7-isomerase), a key enzyme in the cholesterol biosynthesis pathway, which was previously implicated as a target for remyelination. These findings provide valuable insights into the implications for SERMs in remyelination for MS and hormonal research at large.SIGNIFICANCE STATEMENT Therapeutics targeted at remyelination failure, which results in axonal degeneration and ultimately disease progression, represent a large unmet need in the multiple sclerosis (MS) population. Here, we have validated a tolerable European Medicines Agency-approved (U.S. Food and Drug Administration-approved in combination with conjugated estrogens) selective estrogen receptor (ER) modulator (SERM), bazedoxifene (BZA), as a potent agent of oligodendrocyte precursor cell (OPC) differentiation and remyelination. SERMs, which were developed as nuclear ER-α and ER-ß agonists/antagonists, have previously been implicated in remyelination and neuroprotection, following a heavy focus on estrogens with underwhelming and conflicting results. We show that nuclear ERs are not required for SERMs to mediate their potent effects on OPC differentiation and remyelination in vivo and highlight EBP, an enzyme in the cholesterol biosynthesis pathway that could potentially act as a target for SERMs.


Asunto(s)
Indoles/administración & dosificación , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Receptores de Estrógenos/fisiología , Remielinización/efectos de los fármacos , Moduladores Selectivos de los Receptores de Estrógeno/administración & dosificación , Animales , Diferenciación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones Endogámicos C57BL , Esclerosis Múltiple/tratamiento farmacológico , Células Precursoras de Oligodendrocitos/fisiología , Oligodendroglía/fisiología
4.
Cell Rep ; 25(3): 544-550.e3, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30332636

RESUMEN

Emerging evidence suggests that neuronal signaling is important for oligodendrocyte myelination; however, the necessity of this signaling during development is unclear. By eliminating dynamic neuronal signaling along the developing optic nerve, we find that oligodendrocyte differentiation is not dependent on neuronal signaling and that the initiation of myelination is dependent on a permissive substrate, namely supra-threshold axon caliber. Furthermore, we show that loss of dynamic neuronal signaling results in hypermyelination of axons. We propose that oligodendrocyte differentiation is regulated by non-neuronal factors during optic nerve development, whereas myelination is sensitive to the biophysical properties of axonal diameter.


Asunto(s)
Axones/fisiología , Encéfalo/fisiología , Vaina de Mielina/fisiología , Neurogénesis , Oligodendroglía/fisiología , Nervio Óptico/fisiología , Animales , Axones/química , Encéfalo/citología , Diferenciación Celular , Proliferación Celular , Femenino , Masculino , Ratones , Ratones Noqueados , Oligodendroglía/citología , Nervio Óptico/citología , Fosfohidrolasa PTEN/fisiología , Transducción de Señal
5.
Nat Neurosci ; 20(5): 674-680, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28288125

RESUMEN

Regeneration of CNS myelin involves differentiation of oligodendrocytes from oligodendrocyte progenitor cells. In multiple sclerosis, remyelination can fail despite abundant oligodendrocyte progenitor cells, suggesting impairment of oligodendrocyte differentiation. T cells infiltrate the CNS in multiple sclerosis, yet little is known about T cell functions in remyelination. We report that regulatory T cells (Treg) promote oligodendrocyte differentiation and (re)myelination. Treg-deficient mice exhibited substantially impaired remyelination and oligodendrocyte differentiation, which was rescued by adoptive transfer of Treg. In brain slice cultures, Treg accelerated developmental myelination and remyelination, even in the absence of overt inflammation. Treg directly promoted oligodendrocyte progenitor cell differentiation and myelination in vitro. We identified CCN3 as a Treg-derived mediator of oligodendrocyte differentiation and myelination in vitro. These findings reveal a new regenerative function of Treg in the CNS, distinct from immunomodulation. Although the cells were originally named 'Treg' to reflect immunoregulatory roles, this also captures emerging, regenerative Treg functions.


Asunto(s)
Encéfalo/fisiología , Vaina de Mielina/fisiología , Regeneración/fisiología , Linfocitos T Reguladores/fisiología , Animales , Encéfalo/ultraestructura , Diferenciación Celular/fisiología , Femenino , Masculino , Ratones , Proteína Hiperexpresada del Nefroblastoma/fisiología , Oligodendroglía/fisiología , Células Madre/fisiología
6.
J Neurosci ; 36(26): 6937-48, 2016 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-27358452

RESUMEN

UNLABELLED: Myelin controls the time required for an action potential to travel from the neuronal soma to the axon terminal, defining the temporal manner in which information is processed within the CNS. The presence of myelin, the internodal length, and the thickness of the myelin sheath are powerful structural factors that control the velocity and fidelity of action potential transmission. Emerging evidence indicates that myelination is sensitive to environmental experience and neuronal activity. Activity-dependent modulation of myelination can dynamically alter action potential conduction properties but direct functional in vivo evidence and characterization of the underlying myelin changes is lacking. We demonstrate that in mice long-term monocular deprivation increases oligodendrogenesis in the retinogeniculate pathway but shortens myelin internode lengths without affecting other structural properties of myelinated fibers. We also demonstrate that genetically attenuating synaptic glutamate neurotransmission from retinal ganglion cells phenocopies the changes observed after monocular deprivation, suggesting that glutamate may constitute a signal for myelin length regulation. Importantly, we demonstrate that visual deprivation and shortened internodes are associated with a significant reduction in nerve conduction velocity in the optic nerve. Our results reveal the importance of sensory input in the building of myelinated fibers and suggest that this activity-dependent alteration of myelination is important for modifying the conductive properties of brain circuits in response to environmental experience. SIGNIFICANCE STATEMENT: Oligodendrocyte precursor cells differentiate into mature oligodendrocytes and are capable of ensheathing axons with myelin without molecular cues from neurons. However, this default myelination process can be modulated by changes in neuronal activity. Here, we show, for the first time, that experience-dependent activity modifies the length of myelin internodes along axons altering action potential conduction velocity. Such a mechanism would allow for variations in conduction velocities that provide a degree of plasticity in accordance to environmental needs. It will be important in future work to investigate how these changes in myelination and conduction velocity contribute to signal integration in postsynaptic neurons and circuit function.


Asunto(s)
Fibras Nerviosas Mielínicas/fisiología , Conducción Nerviosa/fisiología , Nervio Óptico/fisiología , Visión Monocular/fisiología , Vías Visuales/fisiología , Potenciales de Acción/fisiología , Factores de Edad , Animales , Animales Recién Nacidos , Antígenos/genética , Antígenos/metabolismo , Toxina del Cólera/metabolismo , Cuerpos Geniculados/citología , Cuerpos Geniculados/fisiología , Cuerpos Geniculados/ultraestructura , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vaina de Mielina/metabolismo , Vaina de Mielina/ultraestructura , Fibras Nerviosas Mielínicas/ultraestructura , Conducción Nerviosa/genética , Nervio Óptico/ultraestructura , Organogénesis/genética , Organogénesis/fisiología , Estimulación Luminosa , Proteoglicanos/genética , Proteoglicanos/metabolismo , Células Ganglionares de la Retina/metabolismo , Transmisión Sináptica/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/genética , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo , Vías Visuales/ultraestructura
7.
J Neurosci ; 36(30): 7925-35, 2016 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-27466337

RESUMEN

UNLABELLED: Remyelinating therapies seek to promote restoration of function and normal cellular architecture following demyelination in diseases, such as multiple sclerosis (MS). Functional screening for small molecules or novel targets for remyelination is a major hurdle to the identification and development of rational therapeutics for MS. Recent findings and technical advances provide us with a unique opportunity to provide insight into the cell autonomous mechanisms for remyelination and address this unmet need. Upon screening a G-protein-coupled receptor small-molecule library, we report the identification of a cluster of κ-opioid receptor (KOR) agonists that significantly promotes oligodendrocyte differentiation and myelination. KOR agonists were validated in purified rat oligodendroglial cultures, and the (±)U-50488 compound proved to be most effective for differentiation. (±)U-50488 treatment significantly enhances differentiation and myelination in purified oligodendroglial cocultures and greatly accelerates the kinetics of remyelination in vivo after focal demyelination with lysolecithin. The effect of (±)U-50488 is attenuated by KOR antagonists and completely abolished in KOR-null oligodendroglia. Conditional deletion of KOR in murine oligodendrocyte precursor cells (OPCs) greatly inhibits remyelination after focal demyelination lacking any response to (±)U-50488 treatment. To determine whether agonism of KOR represents a feasible therapeutic approach, human induced pluripotent stem cell-derived OPCs were treated with (±)U-50488. Consistent with findings, differentiation of human OPCs into mature oligodendrocytes was significantly enhanced. Together, KOR is a therapeutic target to consider for future remyelination therapy. SIGNIFICANCE STATEMENT: Remyelination represents a promising strategy to achieve functional recovery in demyelinating diseases, like MS. Thus, identification of potent compounds and targets that promote remyelination represents a critically unmet need. This study reports a cluster of compounds that are highly effective in enhancing remyelination and identifies κ-opioid receptor (KOR) as a positive regulator for oligodendroglial differentiation, implicating KOR agonism as a potential strategy to accelerate remyelination.


Asunto(s)
Enfermedades Desmielinizantes/fisiopatología , Vaina de Mielina/fisiología , Regeneración Nerviosa/fisiología , Oligodendroglía/citología , Oligodendroglía/fisiología , Receptores Opioides kappa/metabolismo , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Enfermedades Desmielinizantes/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Femenino , Masculino , Ratones , Neurogénesis/fisiología , Receptores Opioides kappa/agonistas
8.
Curr Opin Neurobiol ; 39: 47-52, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27128881

RESUMEN

During development oligodendrocyte precursor cells (OPCs) rapidly proliferate and migrate throughout the central nervous system. The mobilization of OPCs is followed by terminal differentiation into mature oligodendrocytes and the subsequent myelination of axons. Differentiation of OPCs is CNS-wide and robust, and yet spatially and temporally restricted. What factors control this precise and coordinated differentiation effort? We discuss evidence for both intrinsic and extrinsic cues in regulating OPC differentiation and gather that extrinsic cues play the leading role in regulating the differentiation of OPCs into mature oligodendrocytes.


Asunto(s)
Diferenciación Celular , Ambiente , Oligodendroglía/citología , Sistema Nervioso Central/citología , Humanos
9.
J Neurosci ; 36(3): 806-13, 2016 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-26791211

RESUMEN

Oligodendrocyte progenitors respond to biophysical or mechanical signals, and it has been reported that mechanostimulation modulates cell proliferation, migration, and differentiation. Here we report the effect of three mechanical stimuli on mouse oligodendrocyte progenitor differentiation and identify the molecular components of the linker of nucleoskeleton and cytoskeleton (LINC) complex (i.e., SYNE1) as transducers of mechanical signals to the nucleus, where they modulate the deposition of repressive histone marks and heterochromatin formation. The expression levels of LINC components increased during progenitor differentiation and silencing the Syne1 gene resulted in aberrant histone marks deposition, chromatin reorganization and impaired myelination. We conclude that spatial constraints, via the actin cytoskeleton and LINC complex, mediate nuclear changes in oligodendrocyte progenitors that favor a default pathway of differentiation. Significance statement: It is recognized that oligodendrocyte progenitors are mechanosensitive cells. However, the molecular mechanisms translating mechanical stimuli into oligodendrocyte differentiation remain elusive. This study identifies components of the mechanotransduction pathway in the oligodendrocyte lineage.


Asunto(s)
Núcleo Celular/metabolismo , Epigénesis Genética/fisiología , Mecanotransducción Celular/fisiología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas Nucleares/biosíntesis , Oligodendroglía/fisiología , Animales , Núcleo Celular/genética , Proteínas del Citoesqueleto , Femenino , Masculino , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética
10.
Nat Commun ; 5: 4991, 2014 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-25255972

RESUMEN

The Schwann cell (SC)-axon interface represents a membrane specialization that integrates axonal signals to coordinate cytoskeletal dynamics resulting in myelination. Here we show that LKB1/Par-4 is asymmetrically localized to the SC-axon interface and co-localizes with the polarity protein Par-3. Using purified SCs and myelinating cocultures, we demonstrate that localization is dependent on the phosphorylation of LKB1 at serine-431. SC-specific deletion of LKB1 significantly attenuates developmental myelination, delaying the initiation and altering the myelin extent into adulthood, resulting in a 30% reduction in the conduction velocity along the adult sciatic nerves. Phosphorylation of LKB1 by protein kinase A is essential to establish the asymmetric localization of LKB1 and Par-3 and rescues the delay in myelination observed in the SC-specific knockout of LKB1. Our findings suggest that SC polarity may coordinate multiple signalling complexes that couple SC-axon contact to the redistribution of specific membrane components necessary to initiate and control myelin extent.


Asunto(s)
Polaridad Celular , Vaina de Mielina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Células de Schwann/citología , Células de Schwann/enzimología , Proteínas Quinasas Activadas por AMP , Proteínas Adaptadoras Transductoras de Señales , Secuencias de Aminoácidos , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Proteínas de Ciclo Celular , Células Cultivadas , Ratones , Ratones Noqueados , Fosforilación , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Ratas , Células de Schwann/metabolismo
11.
Nat Med ; 20(8): 954-960, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24997607

RESUMEN

Functional screening for compounds that promote remyelination represents a major hurdle in the development of rational therapeutics for multiple sclerosis. Screening for remyelination is problematic, as myelination requires the presence of axons. Standard methods do not resolve cell-autonomous effects and are not suited for high-throughput formats. Here we describe a binary indicant for myelination using micropillar arrays (BIMA). Engineered with conical dimensions, micropillars permit resolution of the extent and length of membrane wrapping from a single two-dimensional image. Confocal imaging acquired from the base to the tip of the pillars allows for detection of concentric wrapping observed as 'rings' of myelin. The platform is formatted in 96-well plates, amenable to semiautomated random acquisition and automated detection and quantification. Upon screening 1,000 bioactive molecules, we identified a cluster of antimuscarinic compounds that enhance oligodendrocyte differentiation and remyelination. Our findings demonstrate a new high-throughput screening platform for potential regenerative therapeutics in multiple sclerosis.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Esclerosis Múltiple/tratamiento farmacológico , Antagonistas Muscarínicos/aislamiento & purificación , Fibras Nerviosas Mielínicas/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Clemastina/farmacología , Evaluación Preclínica de Medicamentos/métodos , Femenino , Antagonistas de los Receptores Histamínicos H1/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Antagonistas Muscarínicos/farmacología , Nanoestructuras , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/fisiología , Ratas , Ratas Sprague-Dawley , Regeneración/efectos de los fármacos
12.
Pediatr Res ; 70(1): 15-20, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21436761

RESUMEN

Male sex is an independent risk factor for long-term neurologic deficits in human preterm infants. Using a chronic, sublethal hypoxia (CSH) mouse model of preterm brain injury, we recently demonstrated acute brain volume loss with an increased male susceptibility to hippocampal volume loss and hypomyelination. We now characterize the long-term, sex-specific effects of CSH on cognition and brain growth. Neonatal mice were treated with CSH for 8 d, raised in normoxia thereafter and underwent behavioral testing at 6 wk of age. Behavioral assays sensitive to hippocampal function were chosen. CSH-treated males had impairments in associative learning, spatial memory, and long-term social memory compared with control males. In contrast, CSH-treated females were less impaired. Persistent reductions in hippocampal and cerebellar volumes were found in adult CSH-treated males, whereas regional brain volumes in adult CSH-treated females were indistinguishable from controls. Similar to human preterm infants, males exposed to hypoxia are especially vulnerable to short-term and long-term deficits in cognition and brain growth.


Asunto(s)
Conducta Animal , Encéfalo/patología , Trastornos del Conocimiento/etiología , Cognición , Hipoxia Encefálica/patología , Factores de Edad , Envejecimiento , Animales , Animales Recién Nacidos , Aprendizaje por Asociación , Encéfalo/crecimiento & desarrollo , Proliferación Celular , Enfermedad Crónica , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/fisiopatología , Trastornos del Conocimiento/psicología , Señales (Psicología) , Modelos Animales de Enfermedad , Conducta Exploratoria , Femenino , Hipoxia Encefálica/complicaciones , Hipoxia Encefálica/fisiopatología , Hipoxia Encefálica/psicología , Masculino , Aprendizaje por Laberinto , Memoria , Ratones , Ratones Endogámicos C57BL , Actividad Motora , Tamaño de los Órganos , Castigo , Factores Sexuales , Conducta Social
13.
Pediatr Res ; 66(3): 248-53, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19542909

RESUMEN

Male sex is a well-established risk factor for poor neurodevelopmental outcome after premature birth. The mechanisms behind this sex-related difference are unknown. The damage associated with prematurity can be mimicked in rodents by prolonged exposure to sublethal postnatal hypoxia. This chronic hypoxia leads to anatomical changes in mice that strongly resemble the loss of volume, decreased myelination, and ventriculomegaly seen in preterm newborns. However, no sex differences have been previously noted in this rodent model. We hypothesized that sex comparisons in hypoxic mice would show sex-related differences in brain volume and white matter loss in response to the same degree of hypoxic insult. Mice were placed in chronic sublethal hypoxia from postnatal day 3-11. Cortical, hippocampal, and cerebellar volumes and myelination indices were measured. We found that the male hippocampus, normally larger than the female, undergoes a greater volume loss compared with females (p < 0.05). Myelination, generally greater in males, was significantly disrupted by hypoxia in neonatal male forebrain. These results support the use of this rodent model to investigate the basis of sex-related susceptibility to brain damage and develop new sex-based neuroprotective strategies.


Asunto(s)
Encéfalo/patología , Feto , Hipoxia-Isquemia Encefálica/patología , Caracteres Sexuales , Animales , Animales Recién Nacidos , Encéfalo/anatomía & histología , Encéfalo/fisiología , Proliferación Celular , Femenino , Feto/patología , Feto/fisiología , Humanos , Hipoxia-Isquemia Encefálica/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/metabolismo , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...