Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Brain Behav Immun ; 119: 261-271, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38570102

RESUMEN

Upregulation of soluble tumor necrosis factor (sTNF) cytokine signaling through TNF receptor 1 (TNFR1) and subsequent neuronal hyperexcitability are observed in both animal models and human chronic neuropathic pain (CNP). Previously, we have shown that estrogen modulates sTNF/TNFR1 signaling in CNP, which may contribute to female prevalence of CNP. The estrogen-dependent role of TNFR1-mediated supraspinal neuronal circuitry in CNP remains unknown. In this study, we interrogated the intersect between supraspinal TNFR1 mediated neuronal signaling and sex specificity by selectively removing TNFR1 in Nex + neurons in adult mice (NexCreERT2::TNFR1f/f). We determined that mechanical hypersensitivity induced by chronic constriction injury (CCI) decreases over time in males, but not in females. Subsequently, we investigated two downstream pathways, p38MAPK and NF-κB, important in TNFR1 signaling and injury response. We detected p38MAPK and NF-κB activation in male cortical tissue; however, p38MAPK phosphorylation was reduced in NexCreERT2::TNFR1f/f males. We observed a similar recovery from acute pain in male mice following CCI when p38αMAPK was knocked out of supraspinal Nex + neurons (NexCreERT2::p38αMAPKf/f), while chronic pain developed in female mice. To explore the intersection between estrogen and inflammation in CNP we used a combination therapy of an estrogen receptor ß (ER ß) inhibitor with a sTNF/TNFR1 or general p38MAPK inhibitor. We determined both combination therapies lends therapeutic relief to females following CCI comparable to the response evaluated in male mice. These data suggest that TNFR1/p38αMAPK signaling in Nex + neurons in CNP is male-specific and lack of therapeutic efficacy following sTNF inhibition in females is due to ER ß interference. These studies highlight sex-specific differences in pathways important to pain chronification and elucidate potential therapeutic strategies that would be effective in both sexes.


Asunto(s)
Dolor Crónico , Estrógenos , Neuralgia , Neuronas , Receptores Tipo I de Factores de Necrosis Tumoral , Transducción de Señal , Animales , Neuralgia/metabolismo , Masculino , Femenino , Ratones , Estrógenos/metabolismo , Estrógenos/farmacología , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Neuronas/metabolismo , Dolor Crónico/metabolismo , Transducción de Señal/fisiología , FN-kappa B/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Hiperalgesia/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Factor de Necrosis Tumoral alfa/metabolismo
2.
J Neurosci ; 38(17): 4146-4162, 2018 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-29610439

RESUMEN

Cardiovascular disease and susceptibility to infection are leading causes of morbidity and mortality for individuals with spinal cord injury (SCI). A major contributor to these is autonomic dysreflexia (AD), an amplified reaction of the autonomic nervous system (hallmarked by severe hypertension) in response to sensory stimuli below the injury. Maladaptive plasticity of the spinal sympathetic reflex circuit below the SCI results in AD intensification over time. Mechanisms underlying this maladaptive plasticity are poorly understood, restricting the identification of treatments. Thus, no preventative treatments are currently available. Neuroinflammation has been implicated in other pathologies associated with hyperexcitable neural circuits. Specifically, the soluble form of TNFα (sTNFα) is known to play a role in neuroplasticity. We hypothesize that persistent expression of sTNFα in spinal cord underlies AD exacerbation. To test this, we intrathecally administered XPro1595, a biologic that renders sTNFα nonfunctional, after complete, high-level SCI in female rats. This dramatically attenuated the intensification of colorectal distension-induced and naturally occurring AD events. This improvement is mediated via decreased sprouting of nociceptive primary afferents and activation of the spinal sympathetic reflex circuit. We also examined peripheral vascular function using ex vivo pressurized arterial preparations and immune function via flow cytometric analysis of splenocytes. Diminishing AD via pharmacological inhibition of sTNFα mitigated ensuing vascular hypersensitivity and immune dysfunction. This is the first demonstration that neuroinflammation-induced sTNFα is critical for altering the spinal sympathetic reflex circuit, elucidating a novel mechanism for AD. Importantly, we identify the first potential pharmacological, prophylactic treatment for this life-threatening syndrome.SIGNIFICANCE STATEMENT Autonomic dysreflexia (AD), a disorder that develops after spinal cord injury (SCI) and is hallmarked by sudden, extreme hypertension, contributes to cardiovascular disease and susceptibility to infection, respectively, two leading causes of mortality and morbidity in SCI patients. We demonstrate that neuroinflammation-induced expression of soluble TNFα plays a critical role in AD, elucidating a novel underlying mechanism. We found that intrathecal administration after SCI of a biologic that inhibits soluble TNFα signaling dramatically attenuates AD and significantly reduces AD-associated peripheral vascular and immune dysfunction. We identified mechanisms behind diminished plasticity of neuronal populations within the spinal sympathetic reflex circuit. This study is the first to pinpoint a potential pharmacological, prophylactic strategy to attenuate AD and ensuing cardiovascular and immune dysfunction.


Asunto(s)
Disreflexia Autónoma/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Disreflexia Autónoma/fisiopatología , Células Cultivadas , Femenino , Arterias Mesentéricas/fisiopatología , Ratas , Ratas Wistar , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Médula Espinal/fisiopatología , Bazo/inmunología , Bazo/fisiopatología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/farmacología
3.
J Neuroinflammation ; 11: 159, 2014 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-25204558

RESUMEN

BACKGROUND: Glial cell activation and overproduction of inflammatory mediators in the central nervous system (CNS) have been implicated in acute traumatic injuries to the CNS, including spinal cord injury (SCI). Elevated levels of the proinflammatory cytokine tumor necrosis factor (TNF), which exists in both a soluble (sol) and a transmembrane (tm) form, have been found in the lesioned cord early after injury. The contribution of solTNF versus tmTNF to the development of the lesion is, however, still unclear. METHODS: We tested the effect of systemically or centrally blocking solTNF alone, using XPro1595, versus using the drug etanercept to block both solTNF and tmTNF compared to a placebo vehicle following moderate SCI in mice. Functional outcomes were evaluated using the Basso Mouse Scale, rung walk test, and thermal hyperalgesia analysis. The inflammatory response in the lesioned cord was investigated using immunohistochemistry and western blotting analyses. RESULTS: We found that peripheral administration of anti-TNF therapies had no discernable effect on locomotor performances after SCI. In contrast, central administration of XPro1595 resulted in improved locomotor function, decreased anxiety-related behavior, and reduced damage to the lesioned spinal cord, whereas central administration of etanercept had no therapeutic effects. Improvements in XPro1595-treated mice were accompanied by increases in Toll-like receptor 4 and TNF receptor 2 (TNFR2) protein levels and changes in Iba1 protein expression in microglia/macrophages 7 and 28 days after SCI. CONCLUSIONS: These studies suggest that, by selectively blocking solTNF, XPro1595 is neuroprotective when applied directly to the lesioned cord. This protection may be mediated via alteration of the inflammatory environment without suppression of the neuroprotective effects of tmTNF signaling through TNFR2.


Asunto(s)
Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/patología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Femenino , Bombas de Infusión Implantables , Inyecciones Epidurales , Inyecciones Subcutáneas , Ratones , Traumatismos de la Médula Espinal/metabolismo , Factor de Necrosis Tumoral alfa/administración & dosificación , Factor de Necrosis Tumoral alfa/metabolismo
4.
J Neuroinflammation ; 10: 92, 2013 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-23880092

RESUMEN

BACKGROUND: Astrocytes are taking the center stage in neurotrauma and neurological diseases as they appear to play a dominant role in the inflammatory processes associated with these conditions. Previously, we reported that inhibiting NF-κB activation in astrocytes, using a transgenic mouse model (GFAP-IκBα-dn mice), results in improved functional recovery, increased white matter preservation and axonal sparing following spinal cord injury (SCI). In the present study, we sought to determine whether this improvement, due to inhibiting NF-κB activation in astrocytes, could be the result of enhanced oligodendrogenesis in our transgenic mice. METHODS: To assess oligodendrogenesis in GFAP-IκBα-dn compared to wild-type (WT) littermate mice following SCI, we used bromodeoxyuridine labeling along with cell-specific immuno-histochemistry, confocal microscopy and quantitative cell counts. To further gain insight into the underlying molecular mechanisms leading to increased white matter, we performed a microarray analysis in naïve and 3 days, 3 and 6 weeks following SCI in GFAP-IκBα-dn and WT littermate mice. RESULTS: Inhibition of astroglial NF-κB in GFAP-IκBα-dn mice resulted in enhanced oligodendrogenesis 6 weeks following SCI and was associated with increased levels of myelin proteolipid protein compared to spinal cord injured WT mice. The microarray data showed a large number of differentially expressed genes involved in inflammatory and immune response between WT and transgenic mice. We did not find any difference in the number of microglia/leukocytes infiltrating the spinal cord but did find differences in their level of expression of toll-like receptor 4. We also found increased expression of the chemokine receptor CXCR4 on oligodendrocyte progenitor cells and mature oligodendrocytes in the transgenic mice. Finally TNF receptor 2 levels were significantly higher in the transgenic mice compared to WT following injury. CONCLUSIONS: These studies suggest that one of the beneficial roles of blocking NF-κB in astrocytes is to promote oligodendrogenesis through alteration of the inflammatory environment.


Asunto(s)
Astrocitos/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/fisiología , Neurogénesis/fisiología , Oligodendroglía/fisiología , Traumatismos de la Médula Espinal/metabolismo , Animales , Astrocitos/patología , Femenino , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Transgénicos , Oligodendroglía/patología , Traumatismos de la Médula Espinal/patología , Regulación hacia Arriba/fisiología
5.
Nat Genet ; 31(4): 354-62, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12118253

RESUMEN

Neurofibromatosis type 2 is an autosomal dominant disorder characterized by tumors, predominantly schwannomas, in the nervous system. It is caused by mutations in the gene NF2, encoding the growth regulator schwannomin (also known as merlin). Mutations occur throughout the 17-exon gene, with most resulting in protein truncation and undetectable amounts of schwannomin protein. Pathogenic mutations that result in production of defective schwannomin include in-frame deletions of exon 2 and three independent missense mutations within this same exon. Mice with conditional deletion of exon 2 in Schwann cells develop schwannomas, which confirms the crucial nature of exon 2 for growth control. Here we report that the molecular adaptor paxillin binds directly to schwannomin at residues 50-70, which are encoded by exon 2. This interaction mediates the membrane localization of schwannomin to the plasma membrane, where it associates with beta 1 integrin and erbB2. It defines a pathogenic mechanism for the development of NF2 in humans with mutations in exon 2 of NF2.


Asunto(s)
Membrana Celular/metabolismo , Proteínas del Citoesqueleto/metabolismo , Neurofibromina 2/genética , Neurofibromina 2/metabolismo , Fosfoproteínas/metabolismo , Animales , Sitios de Unión , Células Cultivadas , Exones , Integrina beta1/metabolismo , Ratones , Mutación , Neurofibromatosis 2/genética , Neurofibromatosis 2/fisiopatología , Paxillin , Isoformas de Proteínas , Ratas , Receptor ErbB-2/metabolismo , Células de Schwann/citología , Células de Schwann/metabolismo
6.
Res Sq ; 2023 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-37674712

RESUMEN

Upregulation of soluble tumor necrosis factor (sTNF) cytokine signaling through TNF receptor 1 (TNFR1) and subsequent neuronal hyperexcitability are observed in both animal models and human chronic neuropathic pain (CNP) [1-4]. To test the hypothesis that supraspinal circuitry is critical to pain chronification, we studied the intersect between supraspinal TNFR1 mediated neuronal signaling and sex specificity by selectively removing TNFR1 in Nex + neurons in adult mice (NexCreERT2::TNFR1f/f). We determined that following chronic constriction injury (CCI), pain resolves in males; however, female acute pain transitions to chronic. Subsequently, we investigated two downstream pathways, p38MAPK and NF-κB, important in TNFR1 signaling and injury response. We detected p38αMAPK and NF-κB activation in male cortical tissue; however, p38αMAPK phosphorylation was reduced in NexCreERT2::TNFR1f/f males. We observed similar behavioral results following CCI in NexCreERT2::p38αMAPKf/f mice. Previously, we established estrogen's ability to modulate sTNF/TNFR1 signaling in CNP, which may contribute to female prevalence of CNP [5-9]. To explore the intersection between estrogen and inflammation in CNP we used a combination therapy of an estrogen receptor ß (ER ß) inhibitor with a sTNF/TNFR1 or general p38MAPK inhibitor. We determined both combination therapies lend "male-like" therapeutic relief to females following CCI. These data suggest that TNFR1/p38αMAPK signaling in Nex + neurons in CNP is male-specific and lack of therapeutic efficacy following sTNF inhibition in females is due to ER ß interference. These studies highlight sex-specific differences in pathways important to pain chronification and elucidate potential therapeutic strategies that would be effective in both sexes.

7.
Exp Neurol ; 364: 114394, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37001630

RESUMEN

Postural control is critical for locomotion, allowing for gait changes, obstacle avoidance and navigation of rough terrain. A major problem after spinal cord injury (SCI) is regaining the control of balance to prevent falls and further injury. While the circuits for locomotor pattern generation reside in the spinal cord, postural control consists of multiple, complex networks that interact at the spinal, brainstem and cortical levels. After complete SCI, cortical reorganization establishes novel control of trunk musculature that is required for weight-supported stepping. In this study, we examined the impact of exercise therapy on cortical reorganization in the more clinically relevant models of both moderate and severe midthoracic contusion injury in the rat. Results demonstrate that both spontaneous recovery and therapy induced recovery of weight-supported stepping utilize cortical reorganization. Moreover, exercise therapy further improves outcome by enhancing cortical control of lower thoracic muscles enabling improvements in interlimb coordination associated with improved balance that increases weight-supported stepping. The outcome of this study suggest that cortical control of posture is key to functional improvement in locomotion. This information can be used to improve the timing and type of therapy after SCI by considering changes along the entire neural axis.


Asunto(s)
Contusiones , Traumatismos de la Médula Espinal , Humanos , Ratas , Animales , Locomoción/fisiología , Terapia por Ejercicio/métodos , Marcha/fisiología , Traumatismos de la Médula Espinal/terapia , Músculo Esquelético , Recuperación de la Función
8.
Stem Cells ; 28(7): 1231-42, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20496368

RESUMEN

Ephrins and Eph receptor(s) have recently been implicated in regulating neurogenesis in the adult subventricular zone (SVZ) and rostral migratory stream. Here, we examined the role of ephrinB3-EphB3 signaling in mediating the SVZ response to traumatic brain injury (TBI). Analysis of EphB3 expression showed colocalization with glial fibrillary acidic protein-positive neural stem progenitor cells (NSPCs) and doublecortin-positive neuroblasts, whereas ephrinB3 was expressed outside the neurogenic region. TBI resulted in a significant reduction in EphB3 expression, which coincided with enhanced NSPC survival and proliferation at 3 and 7 days postinjury. Analysis of mice lacking either ephrinB3 (ephrinB3(-/-)) or EphB3 (EphB3(-/-)) showed a significant increase in bromodeoxyuridine (BrdU) incorporation and Ki67 immunoreactivity in the SVZ. Interestingly, cell death was dissimilar between knockout mice, where cell death was reduced in EphB3(-/-) but increased in ephrinB3(-/-) mice. Lateral ventricle infusion of soluble preclustered ephrinB3-Fc reversed the proliferative and cell death defects in ephrinB3(-/-) but not EphB3(-/-) mice and prevented TBI-induced proliferation in wild-type NSPCs. Coincidently, tumor suppressor p53 expression was increased following EphB3 stimulation and is reduced in the absence of either EphB3 or ephrinB3. Furthermore, pharmacological inhibition and siRNA knockdown of p53-attenuated ephrinB3-Fc-mediated growth suppression while having no effect on cell death in cultured NSPCs. These data demonstrate that EphB3 signaling suppresses NSPC proliferation in a p53-dependent manner, induces cell death in the absence of ligand stimulation and is transiently reduced in the SVZ to initiate the expansion and survival of endogenous adult NSPCs following TBI.


Asunto(s)
Lesiones Encefálicas/metabolismo , Ventrículos Cerebrales/metabolismo , Homeostasis , Neuronas/metabolismo , Receptor EphB3/metabolismo , Células Madre/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Envejecimiento , Animales , Lesiones Encefálicas/genética , Lesiones Encefálicas/patología , Proliferación Celular , Células Cultivadas , Ventrículos Cerebrales/citología , Efrina-B3/deficiencia , Efrina-B3/metabolismo , Masculino , Ratones , Ratones Noqueados , Neuronas/citología , ARN Interferente Pequeño/genética , Receptor EphB3/deficiencia , Células Madre/citología , Proteína p53 Supresora de Tumor/genética
9.
Eur J Pain ; 25(4): 801-816, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33296535

RESUMEN

BACKGROUND: The rat mid-thoracic contusion model has been used to study at-level tactile allodynia, a common type of pain that develops after spinal cord injury (SCI). An important advantage of this model is that not all animals develop hypersensitivity. Therefore, it can be used to examine mechanisms that are strictly related to the development of pain-like behaviour separately from mechanisms related to the injury itself. However, how to separate animals that develop hypersensitivity from those that do not is unclear. METHODS: The aims of the current study were to identify where hypersensitivity and spasticity develop and use this information to identify metrics to separate animals that develop hypersensitivity from those that do not to study differences in their behaviour. To accomplish these aims, a grid was used to localize hypersensitivity on the dorsal trunk relative to thoracic dermatomes and supraspinal responses to tactile stimulation were tallied. These supraspinal responses were used to develop a hypersensitivity score to separate animals that develop hypersensitivity, or pain-like response to nonpainful stimuli. RESULTS: Similar to humans, the development of hypersensitivity could occur with the development of spasticity or hyperreflexia. Moreover, the time course and prevalence of hypersensitivity phenotypes (at-, above-, or below level) produced by this model were similar to that observed in humans with SCI. CONCLUSION: However, the amount of spared spinal matter in the cord did not explain the development of hypersensitivity, as previously reported. This approach can be used to study the mechanisms underlying the development of hypersensitivity separately from mechanisms related to injury alone.


Asunto(s)
Contusiones , Traumatismos de la Médula Espinal , Animales , Contusiones/complicaciones , Modelos Animales de Enfermedad , Hiperalgesia/etiología , Ratas , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/complicaciones
10.
Biochim Biophys Acta ; 1793(2): 231-8, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18948148

RESUMEN

Eph receptors have been implicated in regulating a diverse array of cellular functions in the developing nervous system. Recently, Eph receptors have been shown to promote cell death in adult germinal zones; however, their mechanisms of action remain ill-defined. In this study, we demonstrate that EphA4 is a new member of the dependence receptors family, which can initiate cell death in the absence of its ligand ephrinB3. Upon removal of its ligand, EphA4 triggers cell death that is dependent on caspase activation as caspase inhibitors prevent cell death. EphA4 itself is cleaved by caspase-3-like caspase in the intracellular domain at position D773/774, which is necessary for cell death initiation as mutation of the cleavage site abolishes apoptosis. In the adult subventricular zone, abolishing ephrinB3 results in increased cell death, while the absence of EphA4 results in excessive numbers of neuroblasts. Furthermore, infusion of soluble ephrinB3 into the lateral ventricle reduced cell death, and together these results support a dependence role for EphA4 in adult neurogenesis.


Asunto(s)
Apoptosis/efectos de los fármacos , Efrina-B3/farmacología , Neurogénesis/efectos de los fármacos , Receptor EphA4/metabolismo , Animales , Caspasa 3/metabolismo , Línea Celular , Ventrículos Cerebrales/efectos de los fármacos , Ventrículos Cerebrales/enzimología , Ventrículos Cerebrales/patología , Activación Enzimática/efectos de los fármacos , Humanos , Ligandos , Masculino , Ratones , Fosforilación/efectos de los fármacos , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/enzimología , Especificidad por Sustrato/efectos de los fármacos
11.
CNS Neurosci Ther ; 25(8): 884-893, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30941924

RESUMEN

AIM: The activation of the TNFR2 receptor is beneficial in several pathologies of the central nervous system, and this study examines whether it can ameliorate the recovery process following spinal cord injury. METHODS: EHD2-sc-mTNFR2 , an agonist specific for TNFR2, was used to treat neurons exposed to high levels of glutamate in vitro. In vivo, it was infused directly to the spinal cord via osmotic pumps immediately after a contusion to the cord at the T9 level. Locomotion behavior was assessed for 6 weeks, and the tissue was analyzed (lesion size, RNA and protein expression, cell death) after injury. Somatosensory evoked potentials were also measured in response to hindlimb stimulation. RESULTS: The activation of TNFR2 protected neurons from glutamate-mediated excitotoxicity through the activation of phosphoinositide-3 kinase gamma in vitro and improved the locomotion of animals following spinal cord injury. The extent of the injury was not affected by infusing EHD2-sc-mTNFR2 , but higher levels of neurofilament H and 2', 3'-cyclic-nucleotide 3'-phosphodiesterase were observed 6 weeks after the injury. Finally, the activation of TNFR2 after injury increased the neural response recorded in the cortex following hindlimb stimulation. CONCLUSION: The activation of TNFR2 in the spinal cord following contusive injury leads to enhanced locomotion and better cortical responses to hindlimb stimulation.


Asunto(s)
Contusiones/tratamiento farmacológico , Receptores Tipo II del Factor de Necrosis Tumoral/agonistas , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase Ib/fisiología , Citocinas/análisis , Femenino , Ratones , Ratones Endogámicos C57BL , Receptores Tipo II del Factor de Necrosis Tumoral/fisiología , Médula Espinal/efectos de los fármacos , Traumatismos de la Médula Espinal/inmunología
12.
Curr Protoc Stem Cell Biol ; Chapter 2: Unit 2D.8, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22415840

RESUMEN

Endogenous neural stem/progenitor cells (NSPCs) residing in the subventricular zone (SVZ) of the adult mouse forebrain have been shown to enhance their neurogenic potential in response to CNS injury. Mechanisms involved in regulating adult neurogenesis under naïve or stressed conditions can be studied using a monolayer cell-culture system of the nestin-expressing NSPC lineage to analyze proliferation, survival, and differentiation. Here, a protocol for the expansion of NSPCs for studies aimed at understanding the functional role of NSPCs in maintaining adult neurogenic processes is described. This unit outlines detailed procedures for: (1) isolation, maintenance, and culture of the NSPC component of the SVZ niche from the lateral wall of the lateral ventricle; (2) characterization of NSPC functions by examining proliferation, survival, and differentiation; and (3) efficient siRNA transfection methods in 96-well format.


Asunto(s)
Células Madre Adultas/citología , Técnicas de Cultivo de Célula/métodos , Ventrículos Cerebrales/citología , Células-Madre Neurales/citología , Células Madre Adultas/metabolismo , Animales , Adhesión Celular , Diferenciación Celular , Proliferación Celular , Separación Celular , Supervivencia Celular , Células Cultivadas , Masculino , Ratones , Células-Madre Neurales/metabolismo , ARN Interferente Pequeño/metabolismo , Reproducibilidad de los Resultados , Transfección
13.
J Biomol Screen ; 17(6): 785-95, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22492230

RESUMEN

Small-molecule compounds (SMCs) can provide an inexpensive and selective approach to modifying biological responses. High-content analysis (HCA) of SMC libraries can help identify candidate molecules that inhibit or activate cellular responses. In particular, regulation of cell death has important implications for many pathological conditions. Dependence receptors are a new classification of proapoptotic membrane receptors that, unlike classic death receptors, initiate apoptotic signals in the absence of their ligands. EphA4 has recently been identified as a dependence receptor that may have important functions in conditions as disparate as cancer biology and CNS injury and disease. To screen potential candidate SMCs that inhibit or activate EphA4-induced cell death, HCA of an SMC library was performed using stable EphA4-expressing NIH 3T3 cells. Our results describe a high-content method for screening dependence receptor-signaling pathways and demonstrate that several candidate SMCs can inhibit EphA4-mediated cell death.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Receptor EphA4/agonistas , Receptor EphA4/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Apoptosis/efectos de los fármacos , Efrinas/metabolismo , Colorantes Fluorescentes , Humanos , Ratones , Células 3T3 NIH , Receptor EphA4/genética
14.
Int J Dev Neurosci ; 29(1): 9-14, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20969945

RESUMEN

The subventricular zone (SVZ) of the mammalian forebrain is a major source of multipotent stem cells during development, and contributes to neurogenesis throughout the lifespan of the organism. Several studies described molecules regulating adult neurogenesis, however, few of them have examined neurogenesis in the early postnatal period. Adult neurogenesis is regulated in part by ephrinB3 and its receptors, so we examined the role of EphB3 on neural stem/progenitor cell (NSPC) proliferation in early postnatal development in the SVZ. To examine NSPC proliferation, we used BrdU incorporation in both cultured NSPCs and neonatal gene-targeted knockout mice, as well as Ki67 immunostaining in EphB3(-/-) mice. We observed a significant increase in proliferation in cultured NSPCs derived from EphB3(-/-) mice and in the SVZ of EphB3(-/-) mice. These studies support an anti-proliferative role for EphB3 in regulating NSPC numbers in the developing SVZ.


Asunto(s)
Proliferación Celular , Efrina-B3/metabolismo , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Prosencéfalo/citología , Prosencéfalo/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Efrina-B3/genética , Ratones , Ratones Noqueados , Células-Madre Neurales/citología , Receptores de la Familia Eph/genética , Receptores de la Familia Eph/metabolismo
15.
Mol Cell Neurosci ; 31(4): 713-22, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16483793

RESUMEN

Interactions between ephrins and their receptors have been implicated in many processes during central nervous system development. In the adult, ephrins and Eph receptors have been implicated in controlling cell proliferation and neuroblast migration, although there is no direct evidence for the role of ephrinB3 in these functions. In addition, activation of Eph receptors has been shown to regulate transduction pathways important in cell cycle control as well as cell death. We show that ephrinB3 contributes to the control of cell proliferation and survival in the adult subventricular zone (SVZ). EphrinB3(-/-) mice exhibit a significant increase in dividing cells along the lateral ventricle, and altered expression of proteins involved in cell cycle regulation. Gain-of-function approach by infusing soluble ephrinB3-Fc molecules in ephrinB3(-/-) can suppress cell proliferation to wild type levels. At the same time, ephrinB3 also regulates cell survival as greater numbers of cells die in the SVZ of ephrinB3(-/-) mice. Together, our results suggest that ephrinB3 negatively regulates cell cycle progression and cell apoptosis in the adult subventricular zone.


Asunto(s)
Proliferación Celular , Supervivencia Celular , Efrina-B3/metabolismo , Neuronas/fisiología , Receptores de la Familia Eph/metabolismo , Animales , Apoptosis , Encéfalo/anatomía & histología , Encéfalo/metabolismo , Efrina-B1/metabolismo , Efrina-B2/metabolismo , Efrina-B3/genética , Humanos , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso , Neuronas/citología
16.
IUBMB Life ; 56(1): 1-6, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14992373

RESUMEN

Stem cell biology is one of the most exciting, controversial, and debated fields in science today. It has been suggested that neuronal replacement therapy using stem cell transplants may be one possible answer to a host of neuropathological disorders including spinal cord injury, stroke, and neurodegenerative diseases. Important sources for stem cells include the developing embryo and adult central nervous system, but will these populations of cells exhibit similar behavior and responses to stimuli? This review will discuss some important similarities and differences between the embryonic and adult stem cell, as well as the basis for developing therapeutic approaches for stem cell replacement.


Asunto(s)
Neuronas/citología , Células Madre/citología , Adulto , Animales , Diferenciación Celular , División Celular , Movimiento Celular , Senescencia Celular , Sistema Nervioso Central/lesiones , Humanos , Trasplante de Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA