Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Cell Mol Neurobiol ; 36(3): 313-325, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26993508

RESUMEN

Environmental enrichment (EE) consists of increased physical, intellectual, and social activity, and has wide-ranging effects, including enhancing cognition, learning and memory, and motor coordination. Animal studies have demonstrated that EE improves outcome of brain trauma and neurodegenerative disorders, including demyelinating diseases like multiple sclerosis, making it a promising therapeutic option. However, the complexity of applying a robust EE paradigm makes clinical use difficult. A better understanding of the signaling involved in EE-based neuroprotection may allow for development of effective mimetics as an alternative. In prior work, we found that exosomes isolated from the serum of rats exposed to EE impact CNS myelination. Exosomes are naturally occurring nanovesicles containing mRNA, miRNA, and protein, which play important roles in cell function, disease, and immunomodulation. When applied to hippocampal slice cultures or nasally administered to naïve rats, EE-serum exosomes significantly increase myelin content, oligodendrocyte precursor (OPC) and neural stem cell levels, and reduce oxidative stress (OS). We found that rat EE exosomes were enriched in miR-219, which is necessary and sufficient for OPC differentiation into myelinating cells. Thus, peripherally produced exosomes may be a useful therapy for remyelination. Here, we aim to better characterize the impact of EE on CNS health and to determine the cellular source of nutritive exosomes found in serum. We found that exosomes isolated from various circulating immune cell types all increased slice culture myelin content, contained miR-219, and reduced OS, suggesting that EE globally alters immune function in a way that supports brain health.


Asunto(s)
Sistema Nervioso Central/patología , Ambiente , Exosomas/metabolismo , Inflamación/patología , Leucocitos/metabolismo , Vaina de Mielina/metabolismo , Animales , Axones/ultraestructura , Linfocitos B/citología , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Sistema Nervioso Central/metabolismo , Células Dendríticas/citología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Gliosis/patología , Inflamación/genética , Lipopolisacáridos , Ganglios Linfáticos/citología , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Proteína Básica de Mielina/metabolismo , Estrés Oxidativo , Ratas Wistar , Bazo/citología , Linfocitos T/citología
2.
Glia ; 62(2): 284-99, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24339157

RESUMEN

Although commonly considered a disease of white matter, gray matter demyelination is increasingly recognized as an important component of multiple sclerosis (MS) pathogenesis, particularly in the secondary progressive disease phase. Extent of damage to gray matter is strongly correlated to decline in memory and cognitive dysfunction in MS patients. Aging likewise occurs with cognitive decline from myelin loss, and age-associated failure to remyelinate significantly contributes to MS progression. However, recent evidence demonstrates that parabiotic exposure of aged animals to a youthful systemic milieu can promote oligodendrocyte precursor cell (OPC) differentiation and improve remyelination. In the current study, we focus on this potential for stimulating remyelination, and show it involves serum exosomes that increase OPCs and their differentiation into mature myelin-producing cells-both under control conditions and after acute demyelination. Environmental enrichment (EE) of aging animals produced exosomes that mimicked this promyelinating effect. Additionally, stimulating OPC differentiation via exosomes derived from environmentally enriched animals is unlikely to deplete progenitors, as EE itself promotes proliferation of neural stem cells. We found that both young and EE serum-derived exosomes were enriched in miR-219, which is necessary and sufficient for production of myelinating oligodendrocytes by reducing the expression of inhibitory regulators of differentiation. Accordingly, protein transcript levels of these miR-219 target mRNAs decreased following exosome application to slice cultures. Finally, nasal administration of exosomes to aging rats also enhanced myelination. Thus, peripheral circulating cells in young or environmentally enriched animals produce exosomes that may be a useful therapy for remyelination.


Asunto(s)
Sistema Nervioso Central/metabolismo , Exosomas/metabolismo , MicroARNs/metabolismo , Vaina de Mielina/metabolismo , Oligodendroglía/citología , Envejecimiento , Animales , Axones/metabolismo , Axones/patología , Diferenciación Celular/fisiología , Enfermedades Desmielinizantes/metabolismo , Exposición a Riesgos Ambientales , Masculino , Vaina de Mielina/patología , Células-Madre Neurales/citología , Oligodendroglía/metabolismo , Ratas Endogámicas WKY
3.
Glia ; 62(7): 1176-94, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24723305

RESUMEN

Microglia play an important role in fine-tuning neuronal activity. In part, this involves their production of tumor necrosis factor-alpha (TNFα), which increases neuronal excitability. Excessive synaptic activity is necessary to initiate spreading depression (SD). Increased microglial production of proinflammatory cytokines promotes initiation of SD, which, when recurrent, may play a role in conversion of episodic to high frequency and chronic migraine. Previous work shows that this potentiation of SD occurs through increased microglial production of TNFα and reactive oxygen species, both of which are associated with an M1-skewed microglial population. Hence, we explored the role of microglia and their M1 polarization in SD initiation. Selective ablation of microglia from rat hippocampal slice cultures confirmed that microglia are essential for initiation of SD. Application of minocycline to dampen M1 signaling led to increased SD threshold. In addition, we found that SD threshold was increased in rats exposed to environmental enrichment. These rats had increased neocortical levels of interleukin-11 (IL-11), which decreases TNFα signaling and polarized microglia to an M2a-dominant phenotype. M2a microglia reduce proinflammatory signaling and increase production of anti-inflammatory cytokines, and therefore may protect against SD. Nasal administration of IL-11 to mimic effects of environmental enrichment likewise increased M2a polarization and increased SD threshold, an effect also seen in vitro. Similarly, application of conditioned medium from M2a polarized primary microglia to slice cultures also increased SD threshold. Thus, microglia and their polarization state play an essential role in SD initiation, and perhaps by extension migraine with aura and migraine.


Asunto(s)
Ambiente , Hipocampo/fisiología , Potenciales de la Membrana/fisiología , Microglía/fisiología , Neocórtex/fisiología , Transducción de Señal , Animales , Células Cultivadas , Fármacos del Sistema Nervioso Central/farmacología , Citocinas/metabolismo , Hipocampo/efectos de los fármacos , Vivienda para Animales , Interleucina-11/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Microelectrodos , Microglía/efectos de los fármacos , Minociclina/farmacología , Neocórtex/efectos de los fármacos , Estrés Oxidativo/fisiología , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Técnicas de Cultivo de Tejidos , Factor de Necrosis Tumoral alfa/metabolismo
4.
J Neurochem ; 126(5): 662-72, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23586526

RESUMEN

Spreading depression (SD), the most likely cause of migraine aura and perhaps migraine, occurs with increased oxidative stress (OS). SD increases reactive oxygen species (ROS), and ROS, in turn, can signal to increase neuronal excitability,which includes increased SD susceptibility. SD also elevates tumor necrosis factor-α (TNF-α), which increases neuronal excitability. Accordingly, we probed for the cellular origin of OS from SD and its relationship to TNF-α, which might promote SD, using rat hippocampal slice cultures. We observed significantly increased OS from SD in astrocytes and microglia but not in neurons or oligodendrocytes. Since insulin-like growth factor-1 (IGF-1) mitigates OS from SD, we determined the cell types responsible for this effect. We found that IGF-1 significantly decreased microglial but not astrocytic OS from SD. We also show that IGF-1 abrogated the SD-induced TNF-α increase. Furthermore, TNF-α application increased microglial but not astrocytic OS, an effect abrogated by IGF-1. Next,we showed that SD increased SD susceptibility, and does so via TNF-α. This work suggests that microglia promote SD via increased and interrelated ROS and TNF-α signaling. Thus, IGF-1 mitigation of microglial ROS and TNF-α responses maybe targets for novel therapeutics development to prevent SD, and perhaps migraine.


Asunto(s)
Depresión de Propagación Cortical/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Microglía/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Fenómenos Electrofisiológicos , Femenino , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Microscopía Confocal , Neuronas/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Técnicas de Cultivo de Órganos , Embarazo , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Fijación del Tejido , Factor de Necrosis Tumoral alfa/fisiología
5.
J Neurochem ; 122(1): 221-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22524542

RESUMEN

Spreading depression (SD), the likely cause of migraine aura and perhaps migraine, is triggered by widespread and unfettered neuronal hyperexcitability. Migraine and the initiating hyperexcitability of seizure, which involve oxidative stress (OS), are likely interrelated. Environmental enrichment (EE) decreases seizure and can reduce migraine. EE's well-characterized neuroprotective effect involves insulin-like growth factor-1 (IGF-1). Accordingly, we asked if IGF-1 could mitigate the hyperexcitability that initiates SD using rat hippocampal slice cultures. We demonstrate that IGF-1 significantly decreased SD susceptibility and related OS. We mimicked OS of SD and observed that IGF-1 abolished hyperexcitability from OS. Application of an antioxidant significantly decreased SD susceptibility and co-administration of an antioxidant with IGF-1 produced no additive effect, whereas an oxidizer significantly increased SD, and this effect was abrogated by IGF-1. Moreover, IGF-1 significantly decreased baseline OS, despite seemingly paradoxically increasing CA3 bursting. These results suggest that IGF-1 increased endogenous antioxidants to levels sufficient to buffer against the OS of SD. Insulin similarly mitigated SD susceptibility, but required a far greater dose. Since brain IGF-1 increases with EE, and, like insulin, independently functions as an EE mimetic, we suggest that EE mimetics are a novel source of therapeutics for SD, and by extension, migraine.


Asunto(s)
Fenómenos Biofísicos/efectos de los fármacos , Potenciales Evocados/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Estrés Oxidativo/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Análisis de Varianza , Animales , Animales Recién Nacidos , Biofisica , Región CA3 Hipocampal/efectos de los fármacos , Región CA3 Hipocampal/fisiología , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Femenino , Colorantes Fluorescentes/metabolismo , Peróxido de Hidrógeno/farmacología , Insulina/farmacología , Masculino , Técnicas de Cultivo de Órganos , Oxidantes/farmacología , Fosfopiruvato Hidratasa/metabolismo , Ratas , Ratas Wistar , Factores de Tiempo
6.
J Neurochem ; 117(2): 187-96, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21070241

RESUMEN

Cold pre-conditioning reduces subsequent brain injury in small animals but the underlying mechanisms remain undefined. As hypothermia triggers systemic macrophage tumor necrosis factor alpha (TNF-α) production and other neural pre-conditioning stimuli depend on this cytokine, we reasoned that microglia and TNF-α would be similarly involved with cold pre-conditioning neuroprotection. Also, as slice cultures closely approximate their in vivo counterpart and include quiescent microglia, we used rat hippocampal slice cultures to confirm this hypothesis. Furthermore, inflammatory cytokine gene screening with subsequent PCR and immunostaining confirmation of targeted mRNA and related protein changes showed that cold pre-conditioning triggered a significant rise in TNF-α that localized to microglia and a significant rise in interleukin (IL)-11 that localized mainly to hippocampal pyramidal neurons and, more rarely, astrocytes. Importantly, co-stimulation with cold and IL-11, an anti-inflammatory cytokine that inhibits TNF-α expression, abrogated the otherwise evident protection. Instead, cold pre-conditioning coupled with blockade of IL-11 signaling further enhanced neuroprotection from that seen with cold pre-conditioning alone. Thus, physiological activation of brain pro-inflammatory cytokine signaling, and its amplification by inhibition of coincident anti-inflammatory cytokine signaling, may be opportune targets for the development of novel therapeutics that can mimic the protection seen in cold pre-conditioning.


Asunto(s)
Frío , Interleucina-11/antagonistas & inhibidores , Interleucina-11/metabolismo , Neuronas/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Análisis de Varianza , Animales , Animales Recién Nacidos , Anticuerpos/farmacología , Región CA1 Hipocampal/citología , Medio de Cultivo Libre de Suero/farmacología , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Agonistas de Aminoácidos Excitadores/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , N-Metilaspartato/toxicidad , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fosfopiruvato Hidratasa/metabolismo , ARN Mensajero/metabolismo , Ratas , Receptores Tipo I de Factores de Necrosis Tumoral/farmacología , Transducción de Señal/efectos de los fármacos , Temperatura , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Factor de Necrosis Tumoral alfa/genética
7.
Neurosci Lett ; 751: 135809, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33713748

RESUMEN

Migraineurs experience increased oxidative stress which drives the initiation and maintenance of migraine-related pain in animal models and, by extension, migraine in humans. Oxidative stress augments calcitonin gene-related peptide (CGRP) levels, a mediator of migraine pain. Insulin-like growth factor-1 (IGF-1), a neuroprotective growth factor, reduces susceptibility to spreading depression, a preclinical model of migraine, in cultured brain slices by blocking oxidative stress and neuroinflammation from microglia. Similarly, nasal delivery of IGF-1 inhibits spreading depression in vivo. After recurrent cortical spreading depression, nasal administration of IGF-1 also significantly reduces trigeminal ganglion oxidative stress and CGRP levels as well as trigeminocervical c-Fos activation. Here, we probed for the impact of nasal IGF-1 pretreatment on trigeminal system activation using a second well-established preclinical model of migraine, systemic nitroglycerin injection. Adult male rats were treated with one of three doses of IGF-1 (37.5, 75 or 150 µg) and the optimal dose found in males was subsequently used for treatment of female rats. One day later, animals received an intraperitoneal injection of nitroglycerin. Measurements taken two hours later after nitroglycerin alone showed increased surrogate markers of trigeminal activation - oxidative stress and CGRP in the trigeminal ganglion and c-Fos in the trigeminocervical complex compared to vehicle control. These effects were significantly reduced at all doses of IGF-1 for trigeminal ganglion metrics of oxidative stress and CGRP and only at the lowest dose in both males and females for c-Fos. The latter inverted U-shaped or hormetic response is seen in enzyme-targeting drugs. While the specific mechanisms remain to be explored, our data here supports the ability of IGF-1 to preserve mitochondrial and antioxidant pathway homeostasis as means to prevent nociceptive activation in the trigeminal system produced by an experimental migraine model.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Trastornos Migrañosos/tratamiento farmacológico , Nitroglicerina/farmacología , Estrés Oxidativo , Ganglio del Trigémino/metabolismo , Administración Intranasal , Animales , Femenino , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Masculino , Trastornos Migrañosos/etiología , Trastornos Migrañosos/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Wistar , Ganglio del Trigémino/efectos de los fármacos , Ganglio del Trigémino/fisiología
8.
PLoS One ; 16(8): e0255778, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34388189

RESUMEN

Extracellular vesicles secreted from IFNγ-stimulated rat dendritic cells (referred to here as IFNγ-DC-EVs) contain miRNAs which promote myelination (including but not limited to miR-219), and preferentially enter oligodendrocytes in brain slice cultures. IFNγ-DC-EVs also increase myelination when nasally administered to naïve rats. While we can infer that these extracellular vesicles enter the CNS from functional studies, here we demonstrate biodistribution throughout the brain after nasal delivery by way of imaging studies. After nasal administration, Xenolight DiR-labelled IFNγ-DC-EVs were detected 30 minutes later throughout the brain and the cervical spinal cord. We next examined cellular uptake of IFNγ-DC-EVs by transfecting IFNγ-DC-EVs with mCherry mRNA prior to nasal administration. mCherry-positive cells were found along the rostrocaudal axis of the brain to the brainstem. These cells morphologically resembled oligodendrocytes, and indeed cell-specific co-staining for neurons, astrocytes, microglia and oligodendrocytes showed that mcherry positive cells were predominantly oligodendrocytes. This is in keeping with our prior in vitro results showing that IFNγ-DC-EVs are preferentially taken up by oligodendrocytes, and to a lesser extent, microglia. To confirm that IFNγ-DC-EVs delivered cargo to oligodendrocytes, we quantified protein levels of miR-219 mRNA targets expressed in oligodendrocyte lineage cells, and found significantly reduced expression. Finally, we compared intranasal versus intravenous delivery of Xenolight DiR-labelled IFNγ-DC-EVs. Though labelled IFNγ-DC-EVs entered the CNS via both routes, we found that nasal delivery more specifically targeted the CNS with less accumulation in the liver. Taken together, these data show that intranasal administration is an effective route for delivery of IFNγ-DC-EVs to the CNS, and provides additional support for their development as an EV-based neurotherapeutic that, for the first time, targets oligodendrocytes.


Asunto(s)
Células Dendríticas/química , Vesículas Extracelulares/química , Interferón gamma/farmacología , MicroARNs/genética , Administración Intranasal , Administración Intravenosa , Animales , Astrocitos/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Médula Cervical/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Interferón gamma/química , Microglía/efectos de los fármacos , Vaina de Mielina/genética , Neuronas/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Ratas
9.
Brain Res ; 1732: 146673, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31978377

RESUMEN

Migraineurs can show brain hyperexcitability and oxidative stress that may promote headache. Since hyperexcitability can enhance oxidative stress which promotes hyperexcitability, ending this feed-back loop may reduce migraine. Neocortical spreading depression, an animal model of migraine begins with hyperexcitability and triggers oxidative stress in the neocortical area involved and in the trigeminal system, which is important to pain pathway nociceptive activation in migraine. Additionally, oxidative stress causes increased trigeminal ganglion calcitonin gene-related peptide release and oxidative stress can reduce spreading depression threshold. Insulin-like growth factor-1 significantly protects against spreading depression in vitro by reducing oxidative stress and it is effective against spreading depression after intranasal delivery to animals. Here, we used adult male rats and extend this work to study the trigeminal system where insulin-like growth factor-1 receptors are highly expressed. Recurrent neocortical spreading depression significantly increased surrogate markers of trigeminal activation - immunostaining for trigeminal ganglion oxidative stress, calcitonin gene related peptide levels and c-fos in the trigeminocervical complex versus sham. These effects were significantly reduced by intranasal delivery of insulin-like growth factor-1 a day before recurrent neocortical spreading depression. Furthermore, intranasal treatment with insulin-like growth factor-1 significantly reduced naïve levels of trigeminal ganglion calcitonin gene related peptide versus sham with no impact on blood glucose levels. Intranasal delivery of insulin-like growth factor-1 not only mitigates neocortical spreading depression, a cause of migraine hyperexcitability modeled in animals, but also when neocortical spreading depression is triggered by supra-threshold stimuli, insulin-like growth factor-1 effectively reduces nociceptive activation in the trigeminal system.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/metabolismo , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Neuronas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ganglio del Trigémino/efectos de los fármacos , Animales , Neuronas/metabolismo , Ratas , Receptores de Péptido Relacionado con el Gen de Calcitonina/metabolismo , Ganglio del Trigémino/metabolismo
10.
J Neurosci ; 28(47): 12199-211, 2008 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-19020014

RESUMEN

In brain, monomeric immunoglobin G (IgG) is regarded as quiescent and only poised to initiate potentially injurious inflammatory reactions via immune complex formation associated with phagocytosis and tumor necrosis factor alpha (TNF-alpha) production in response to disease. Using rat hippocampal slice and microglial cultures, here we show instead that physiological levels (i.e., 0.2-20 microg/ml) of monomeric IgG unassociated with disease triggered benign low-level proinflammatory signaling that was neuroprotective against CA1 area excitotoxicity and followed a U-shaped or hormetic dose-response. The data indicate that physiological IgG levels activated microglia by enhancing recycling endocytosis plus TNF-alpha release from these cells to produce the neuroprotection. Minocycline, known for its anti-inflammatory and neuroprotective effects when given after disease onset, abrogated IgG-mediated neuroprotection and related microglial effects when given before injury. In contrast, E-prostanoid receptor subtype 2 (EP2) activation, which served as an exemplary paracrine stimulus like the one expected from neuronal activity, amplified IgG-mediated increased microglial recycling endocytosis and TNF-alpha production. Furthermore, like monomeric IgG these EP2 related effects took days to be effective, suggesting both were adaptive anabolic effects consistent with those seen from other long-term preconditioning stimuli requiring de novo protein synthesis. The data provide the first evidence that brain monomeric IgG at physiological levels can have signaling function via enhanced recycling endocytosis/TNF-alpha production from microglia unassociated with disease and that these IgG-mediated changes may be a means by which paracrine signaling from neuronal activity influences microglia to evoke neuroprotection. The data provide further support that low-level proinflammatory neural immune signaling unassociated with disease enhances brain function.


Asunto(s)
Endocitosis/fisiología , Inmunoglobulina G/farmacología , Microglía/efectos de los fármacos , Microglía/fisiología , Fármacos Neuroprotectores/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Células Cultivadas , Dinoprostona/farmacología , Relación Dosis-Respuesta a Droga , Ectodisplasinas/metabolismo , Endocitosis/efectos de los fármacos , Glucosa/deficiencia , Hipocampo , Hipoxia , Inmunoglobulina G/líquido cefalorraquídeo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Minociclina/farmacología , N-Metilaspartato/toxicidad , Neurotoxinas/toxicidad , Técnicas de Cultivo de Órganos , Fosfopiruvato Hidratasa/metabolismo , Ratas , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/farmacología , Proteínas de Unión al GTP rab/metabolismo
11.
Am J Pathol ; 173(5): 1508-17, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18818381

RESUMEN

The T-cell-derived, pleiotropic cytokine interferon (IFN)-gamma is believed to play a key regulatory role in immune-mediated demyelinating disorders of the central nervous system, including multiple sclerosis and experimental autoimmune encephalomyelitis. Our previous work has demonstrated that the endoplasmic reticulum (ER) stress response modulates the response of oligodendrocytes to this cytokine. The ER stress response activates the pancreatic ER kinase, which coordinates an adaptive program known as the integrated stress response by phosphorylating translation initiation factor 2alpha (eIF2alpha). In this study, we found that growth arrest and DNA damage 34 (GADD34), a stress-inducible regulatory subunit of a phosphatase complex that dephosphorylates eIF2alpha, was selectively up-regulated in myelinating oligodendrocytes in mice that ectopically expressed IFN-gamma in the central nervous system. We also found that a GADD34 mutant strain of mice displayed increased levels of phosphorylated eIF2alpha (p-eIF2alpha) in myelinating oligodendrocytes when exposure to IFN-gamma, as well as diminished oligodendrocyte loss and hypomyelination. Furthermore, treatment with salubrinal, a small chemical compound that specifically inhibits protein phosphatase 1(PP1)-GADD34 phosphatase activity, increased the levels of p-eIF2alpha and ameliorated hypomyelination and oligodendrocyte loss in cultured hippocampal slices exposed to IFN-gamma. Thus, our data provide evidence that an enhanced integrated stress response could promote oligodendrocyte survival in immune-mediated demyelination diseases.


Asunto(s)
Interferón gamma/farmacología , Vaina de Mielina/patología , Oligodendroglía/patología , Estrés Fisiológico/patología , Animales , Antígenos de Diferenciación/metabolismo , Proteínas de Ciclo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Cinamatos/farmacología , Factor 2 Eucariótico de Iniciación/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/ultraestructura , Inmunidad/efectos de los fármacos , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Fenotipo , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 1 , Ratas , Tiourea/análogos & derivados , Tiourea/farmacología
12.
Front Neurosci ; 13: 942, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31551696

RESUMEN

Migraine is a common headache disorder characterized by unilateral, intense headaches. In migraine with aura, the painful headache is preceded by focal neurological symptoms that can be visual, sensory, or motor in nature. Spreading depression (the most likely cause of migraine with aura and perhaps related headache pain) results in increased neuronal excitability and related increases in inflammation and production of reactive oxygen species. This in turn can promote the transformation of low-frequency, episodic migraine into higher-frequency and eventually chronic migraine. Though migraine affects 11% of adults worldwide, with 3% experiencing chronic headache, existing therapies offer only modest benefits. Here, we focus on the mechanisms by which environmental enrichment (i.e., volitionally increased intellectual, social, and physical activity) mitigates spreading depression. In prior work, we have shown that exposure to environmental enrichment reduces susceptibility to spreading depression in rats. This protective effect is at least in part due to environmental enrichment-mediated changes in the character of serum exosomes produced by circulating immune cells. We went on to show that environmental enrichment-mimetic exosomes can be produced by stimulating dendritic cells with low levels of interferon gamma (a cytokine that is phasically increased during environmental enrichment). Interferon gamma-stimulated dendritic cell exosomes (IFNγ-DC-Exos) significantly improve myelination and reduce oxidative stress when applied to hippocampal slice cultures. Here, we propose that they may also be effective against spreading depression. We found that administration of IFNγ-DC-Exos reduced susceptibility to spreading depression in vivo and in vitro, suggesting that IFNγ-DC-Exos may be a potential therapeutic for migraine.

13.
Brain Res ; 1677: 47-57, 2017 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-28951235

RESUMEN

Spreading depression (SD) is a wave of cellular depolarization that travels slowly through susceptible gray matter brain areas. SD is the most likely cause of migraine aura and perhaps migraine pain, and is a well-accepted animal model of migraine. Identification of therapeutics that can prevent SD may have clinical relevance toward migraine treatment. Here we show that insulin-like growth factor-1 (IGF-1) significantly inhibited neocortical SD in vivo after intranasal delivery to rats. A single dose of IGF-1 inhibited SD within an hour, and continued to protect for at least seven days thereafter. A two-week course of IGF-1, administered every third day, further decreased SD susceptibility and showed no aberrant effects on glial activation, nasal mucosa, or serum markers of toxicity. SD begets SD in vitro by mechanisms that involve microglial activation. We add to this relationship by showing that recurrent SD in vivo increased susceptibility to subsequent SD, and that intervention with IGF-1 significantly interrupted this pathology. These findings support nasal administration of IGF-1 as a novel intervention capable of mitigating SD susceptibility, and as a result, potentially migraine.


Asunto(s)
Depresión de Propagación Cortical/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Neocórtex/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Administración Intranasal , Animales , Depresión de Propagación Cortical/fisiología , Humanos , Masculino , Mucosa Nasal/citología , Mucosa Nasal/efectos de los fármacos , Neocórtex/citología , Neocórtex/fisiología , Neuroglía/citología , Neuroglía/efectos de los fármacos , Cloruro de Potasio , Distribución Aleatoria , Ratas Wistar
14.
Fluids Barriers CNS ; 14(1): 12, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28457227

RESUMEN

A trans-agency workshop on the blood-brain interface (BBI), sponsored by the National Heart, Lung and Blood Institute, the National Cancer Institute and the Combat Casualty Care Research Program at the Department of Defense, was conducted in Bethesda MD on June 7-8, 2016. The workshop was structured into four sessions: (1) blood sciences; (2) exosome therapeutics; (3) next generation in vitro blood-brain barrier (BBB) models; and (4) BBB delivery and targeting. The first day of the workshop focused on the physiology of the blood and neuro-vascular unit, blood or biofluid-based molecular markers, extracellular vesicles associated with brain injury, and how these entities can be employed to better evaluate injury states and/or deliver therapeutics. The second day of the workshop focused on technical advances in in vitro models, BBB manipulations and nanoparticle-based drug carrier designs, with the goal of improving drug delivery to the central nervous system. The presentations and discussions underscored the role of the BBI in brain injury, as well as the role of the BBB as both a limiting factor and a potential conduit for drug delivery to the brain. At the conclusion of the meeting, the participants discussed challenges and opportunities confronting BBI translational researchers. In particular, the participants recommended using BBI translational research to stimulate advances in diagnostics, as well as targeted delivery approaches for detection and therapy of both brain injury and disease.


Asunto(s)
Barrera Hematoencefálica/fisiopatología , Encefalopatías/patología , National Institutes of Health (U.S.) , Investigación Biomédica Traslacional , Animales , Transporte Biológico , Barrera Hematoencefálica/diagnóstico por imagen , Barrera Hematoencefálica/patología , Encefalopatías/diagnóstico por imagen , Encefalopatías/fisiopatología , Humanos , Imagen por Resonancia Magnética , Estados Unidos
15.
J Neurosci ; 25(15): 3952-61, 2005 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-15829647

RESUMEN

Spreading depression (SD) involves current flow through principal neurons, but the pattern of current flow over the expanse of susceptible tissues or individual principal neurons remains undefined. Accordingly, tissue and single cell maps made from digital imaging of voltage-sensitive dye changes in hippocampal organotypic cultures undergoing SD were processed via optical current source density analysis to reveal the currents associated with pyramidal neurons. Two distinctive current flow patterns were seen. The first was a trilaminar pattern (420 microm2) that developed with the onset of SD in CA3 pyramidal neurons, in which SD most often began. This initial pattern comprised a somatic current sink with current sources to either side in the dendrites that lasted for seconds extending into the first aspect of the classical "inverted saddle" interstitial direct current waveform of SD. Next, the somatic sink backpropagated at a speed of millimeters per minute into the proximal dendrites, resulting in a reversal of the initial current flow pattern to its second orientation, namely dendritic sinks associated with a somatic source. The latter persisted for the remainder of SD in CA3 and was the only pattern seen in CA1, in which SD was rarely initiated. This backpropagating SD current flow resembles that of activity-dependent synaptic activation. Retrograde and associative signaling via principal neuron current flow is a key means to affect tissue function, including synaptic activation and, by extension, perhaps SD. Such current-related postsynaptic signaling might not only help explain SD but also neuroprotection and migraine, two phenomena increasingly recognized as being related to SD.


Asunto(s)
Depresión de Propagación Cortical/fisiología , Hipocampo/citología , Células Piramidales/fisiología , Animales , Animales Recién Nacidos , Diagnóstico por Imagen/métodos , Estimulación Eléctrica/métodos , Inmunohistoquímica/métodos , Lisina/análogos & derivados , Lisina/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Potenciales de la Membrana/efectos de la radiación , Modelos Neurológicos , Proteínas de Neurofilamentos/metabolismo , Técnicas de Cultivo de Órganos , Técnicas de Placa-Clamp/métodos , Ratas , Ratas Wistar , Estadísticas no Paramétricas , Estirenos/metabolismo , Factores de Tiempo
16.
J Interferon Cytokine Res ; 35(10): 795-807, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26083947

RESUMEN

The detrimental effects of T-cell-secreted interferon gamma (IFNγ) on oxidative stress (OS) and demyelination in multiple sclerosis (MS) are well recognized. Recently, we demonstrated that IFNγ-mediated damage to myelin also increases susceptibility to spreading depression (SD; the likely basis of migraine with aura). However, before onset of MS, induction of physiological levels of IFNγ, like that produced by environmental enrichment (EE), protects against demyelination and OS. Accordingly, we focused on the potential for physiological levels of IFNγ to protect against SD. EE, which occurs with a moderate and phasic increase in proinflammatory cytokines, reduces migraine frequency. Thus, we applied phasic or pulsed IFNγ to brain slice cultures to emulate EE. This treatment reduced OS, increased myelin basic protein, a marker for myelin, and reduced susceptibility to SD. Building on our research on exosomes in EE-based neuroprotection, we found that IFNγ stimulation of slice cultures induced release of exosomes, likely from the microglia that produce the same protective effects as IFNγ treatment when applied to naive cultures. Finally, nasal administration of IFNγ to rats recapitulated in vitro effects, reducing OS, increasing myelin, and reducing SD. These results support phasic IFNγ signaling as a therapeutic target for prevention of SD and, by extension, migraine.


Asunto(s)
Depresión/metabolismo , Exosomas/metabolismo , Interferón gamma/farmacología , Animales , Células Cultivadas , Depresión/tratamiento farmacológico , Depresión/genética , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Glutatión/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Interferón gamma/administración & dosificación , MicroARNs/genética , Microglía/efectos de los fármacos , Microglía/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Técnicas de Cultivo de Tejidos
17.
Exp Neurol ; 264: 43-54, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25500111

RESUMEN

Multiple sclerosis and migraine with aura are clinically correlated and both show imaging changes suggestive of myelin disruption. Furthermore, cortical myelin loss in the cuprizone animal model of multiple sclerosis enhances susceptibility to spreading depression, the likely underlying cause of migraine with aura. Since multiple sclerosis pathology involves inflammatory T cell lymphocyte production of interferon-gamma and a resulting increase in oxidative stress, we tested the hypothesis that spreading depression disrupts myelin through similar signaling pathways. Rat hippocampal slice cultures were initially used to explore myelin loss in spreading depression, since they contain T cells, and allow for controlled tissue microenvironment. These experiments were then translated to the in vivo condition in neocortex. Spreading depression in slice cultures induced significant loss of myelin integrity and myelin basic protein one day later, with gradual recovery by seven days. Myelin basic protein loss was abrogated by T cell depletion, neutralization of interferon-gamma, and pharmacological inhibition of neutral sphingomyelinase-2. Conversely, one day after exposure to interferon-gamma, significant reductions in spreading depression threshold, increases in oxidative stress, and reduced levels of glutathione, an endogenous neutral sphingomyelinase-2 inhibitor, emerged. Similarly, spreading depression triggered significant T cell accumulation, sphingomyelinase activation, increased oxidative stress, and reduction of gray and white matter myelin in vivo. Myelin disruption is involved in spreading depression, thereby providing pathophysiological links between multiple sclerosis and migraine with aura. Myelin disruption may promote spreading depression by enhancing aberrant excitability. Thus, preservation of myelin integrity may provide novel therapeutic targets for migraine with aura.


Asunto(s)
Depresión de Propagación Cortical/fisiología , Interferón gamma/metabolismo , Vaina de Mielina/metabolismo , Neocórtex/fisiología , Transducción de Señal/fisiología , Compuestos de Anilina/farmacología , Animales , Anticuerpos/farmacología , Compuestos de Bencilideno/farmacología , Citocinas/inmunología , Citocinas/metabolismo , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Hipocampo/fisiología , Técnicas In Vitro , Interferón gamma/inmunología , Masculino , Proteína Básica de Mielina/metabolismo , Vaina de Mielina/ultraestructura , Neocórtex/ultraestructura , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Esfingomielina Fosfodiesterasa/metabolismo , Linfocitos T/metabolismo
18.
J Extracell Vesicles ; 4: 27575, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26320942

RESUMEN

The NIH Extracellular RNA Communication Program's initiative on clinical utility of extracellular RNAs and therapeutic agents and developing scalable technologies is reviewed here. Background information and details of the projects are presented. The work has focused on modulation of target cell fate by extracellular vesicles (EVs) and RNA. Work on plant-derived vesicles is of intense interest, and non-mammalian sources of vesicles may represent a very promising source for different therapeutic approaches. Retro-viral-like particles are intriguing. Clearly, EVs share pathways with the assembly machinery of several other viruses, including human endogenous retrovirals (HERVs), and this convergence may explain the observation of viral-like particles containing viral proteins and nucleic acid in EVs. Dramatic effect on regeneration of damaged bone marrow, renal, pulmonary and cardiovascular tissue is demonstrated and discussed. These studies show restoration of injured cell function and the importance of heterogeneity of different vesicle populations. The potential for neural regeneration is explored, and the capacity to promote and reverse neoplasia by EV exposure is described. The tremendous clinical potential of EVs underlies many of these projects, and the importance of regulatory issues and the necessity of general manufacturing production (GMP) studies for eventual clinical trials are emphasized. Clinical trials are already being pursued and should expand dramatically in the near future.

19.
J Cereb Blood Flow Metab ; 24(8): 829-39, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15362713

RESUMEN

Cytokines are involved in ischemic tolerance, including that triggered by spreading depression (SD), yet their roles in neuroprotection remain incompletely defined. The latter may stem from the pleiotropic nature of these signaling molecules whose complexities for interaction might be better deciphered through simultaneous measurement of multiple targeted proteins. Accordingly, the authors used microsphere-based flow cytometric immunoassays and hippocampal organotypic cultures (HOTCs) to characterize the magnitude, time course, and diversity of cytokine (interleukin [IL] 1alpha, IL-1beta, IL-2, IL-4, IL-6, IL-10, granulocyte-macrophage colony-stimulating factor [GM-CSF], interferon-gamma [IFN-gamma], and tumor necrosis factor-alpha [TNF-alpha]) response to SD. GM-CSF was not detected in HOTCs or media. However, SD triggered a significant, generalized increase in seven cytokines evident in HOTCs 6 hours later, with the remaining cytokine, IL-1beta, becoming significantly different at 1 and 3 days. Additionally, these changes extended to include surrounding media for IL-6 and TNF-alpha by 1 and 3 days. This increase was localized to microglia via immunostaining for IL-1alpha, IL-1beta, and interferon-y. IL-10, although significantly more abundant in HOTCs 6 hours after SD, was significantly less abundant in surrounding media at that time and at 1 day. Finally, the generalized early increase in tissue cytokines later settled to a pattern at 3 days of recovery centering on changes in IL-1alpha, IL-1beta, and TNF-alpha, cytokines capable of modulating ischemic injury.


Asunto(s)
Depresión de Propagación Cortical/fisiología , Citocinas/biosíntesis , Hipocampo/fisiología , Animales , Estimulación Eléctrica , Electrofisiología , Citometría de Flujo , Inmunoensayo , Inmunohistoquímica , Microesferas , Técnicas de Cultivo de Órganos
20.
Brain Res ; 998(2): 155-63, 2004 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-14751586

RESUMEN

Neurons preserve stable electrophysiological properties despite ongoing changes in morphology and connectivity throughout their lifetime. This dynamic compensatory adjustment, termed 'homeostatic plasticity', may be a fundamental means by which the brain normalizes its excitability, and is possibly altered in disease states such as epilepsy. Despite this significance, the cellular mechanisms of homeostatic plasticity are incompletely understood. Using field potential analyses, we observed a compensatory enhancement of neural excitability after 48 h of activity deprivation via tetrodotoxin (TTX) in hippocampal slice cultures. Because activity deprivation can enhance voltage-gated sodium channel (VGSC) currents, we used Western blot analyses to probe for these channels in control and activity-deprived slice cultures. A significant upregulation of VGSCs expression was evident after activity deprivation. Furthermore, immunohistochemistry revealed this upregulation to occur along primarily pyramidal cell dendrites. Western blot analyses of cultures after 1 day of recovery from activity deprivation showed that VGSC levels returned to control levels, indicating that multiple molecular mechanisms contribute to enhanced excitability. Because of their longevity and in vivo-like cytoarchitecture, we conclude that slice cultures may be highly useful for investigating homeostatic plasticity. Furthermore, we demonstrate that enhanced excitability involves changes in channel expression with a targeted localization likely profound transform the integrative capacities of hippocampal pyramidal cells and their dendrites.


Asunto(s)
Hipocampo/fisiología , Red Nerviosa/fisiología , Plasticidad Neuronal/fisiología , Canales de Potasio con Entrada de Voltaje/biosíntesis , Anestésicos Locales/farmacología , Animales , Western Blotting , Electrofisiología , Hipocampo/efectos de los fármacos , Homeostasis/fisiología , Inmunohistoquímica , Plasticidad Neuronal/efectos de los fármacos , Técnicas de Cultivo de Órganos , Canales de Potasio con Entrada de Voltaje/efectos de los fármacos , Ratas , Ratas Wistar , Tetrodotoxina/farmacología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA