Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 115(21): 5528-5533, 2018 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-29728463

RESUMEN

Although immune attack against central nervous system (CNS) myelin is a central feature of multiple sclerosis (MS), its root cause is unresolved. In this report, we provide direct evidence that subtle biochemical modifications to brain myelin elicit pathological immune responses with radiological and histological properties similar to MS lesions. A subtle myelinopathy induced by abbreviated cuprizone treatment, coupled with subsequent immune stimulation, resulted in lesions of inflammatory demyelination. The degree of myelin injury dictated the resulting immune response; biochemical damage that was too limited or too extensive failed to trigger overt pathology. An inhibitor of peptidyl arginine deiminases (PADs), enzymes that alter myelin structure and correlate with MS lesion severity, mitigated pathology even when administered only during the myelin-altering phase. Moreover, cultured splenocytes were reactive against donor myelin isolates, a response that was substantially muted when splenocytes were exposed to myelin from donors treated with PAD inhibitors. By showing that a primary biochemical myelinopathy can trigger secondary pathological inflammation, "cuprizone autoimmune encephalitis" potentially reconciles conflicting theories about MS pathogenesis and provides a strong rationale for investigating myelin as a primary target for early, preventative therapy.


Asunto(s)
Enfermedades Desmielinizantes/etiología , Modelos Animales de Enfermedad , Encefalitis/patología , Enfermedad de Hashimoto/patología , Inflamación/patología , Esclerosis Múltiple/etiología , Vaina de Mielina/patología , Animales , Cuprizona/toxicidad , Enfermedades Desmielinizantes/patología , Encefalitis/inducido químicamente , Encefalitis/inmunología , Enfermedad de Hashimoto/inducido químicamente , Enfermedad de Hashimoto/inmunología , Humanos , Hidrolasas/genética , Hidrolasas/metabolismo , Inflamación/inducido químicamente , Inflamación/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Inhibidores de la Monoaminooxidasa/toxicidad , Esclerosis Múltiple/patología , Vaina de Mielina/inmunología , Vaina de Mielina/metabolismo
2.
Proc Natl Acad Sci U S A ; 109(5): 1737-42, 2012 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-22307640

RESUMEN

N-methyl-d-aspartate receptors (NMDARs) mediate critical CNS functions, whereas excessive activity contributes to neuronal damage. At physiological glycine concentrations, NMDAR currents recorded from cultured rodent hippocampal neurons exhibited strong desensitization in the continued presence of NMDA, thus protecting neurons from calcium overload. Reducing copper availability by specific chelators (bathocuproine disulfonate, cuprizone) induced nondesensitizing NMDAR currents even at physiologically low glycine concentrations. This effect was mimicked by, and was not additive with, genetic ablation of cellular prion protein (PrP(C)), a key copper-binding protein in the CNS. Acute ablation of PrP(C) by enzymatically cleaving its cell-surface GPI anchor yielded similar effects. Biochemical studies and electrophysiological measurements revealed that PrP(C) interacts with the NMDAR complex in a copper-dependent manner to allosterically reduce glycine affinity for the receptor. Synthetic human Aß(1-42) (10 nM-5 µM) produced an identical effect that could be mitigated by addition of excess copper ions or NMDAR blockers. Taken together, Aß(1-42), copper chelators, or PrP(C) inactivation all enhance the activity of glycine at the NMDAR, giving rise to pathologically large nondesensitizing steady-state NMDAR currents and neurotoxicity. We propose a physiological role for PrP(C), one that limits excessive NMDAR activity that might otherwise promote neuronal damage. In addition, we provide a unifying molecular mechanism whereby toxic species of Aß(1-42) might mediate neuronal and synaptic injury, at least in part, by disrupting the normal copper-mediated, PrP(C)-dependent inhibition of excessive activity of this highly calcium-permeable glutamate receptor.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Cobre/metabolismo , Fragmentos de Péptidos/toxicidad , Proteínas PrPC/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Células Cultivadas , Femenino , Cinética , Embarazo , Ratas , Ratas Sprague-Dawley
3.
Front Cell Neurosci ; 18: 1426231, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39161786

RESUMEN

Multiple sclerosis (MS) is a frequently disabling neurological disorder characterized by symptoms, clinical signs and imaging abnormalities that typically fluctuate over time, affecting any level of the CNS. Prominent lymphocytic inflammation, many genetic susceptibility variants involving immune pathways, as well as potent responses of the neuroinflammatory component to immunomodulating drugs, have led to the natural conclusion that this disease is driven by a primary autoimmune process. In this Hypothesis and Theory article, we discuss emerging data that cast doubt on this assumption. After three decades of therapeutic experience, what has become clear is that potent immune modulators are highly effective at suppressing inflammatory relapses, yet exhibit very limited effects on the later progressive phase of MS. Moreover, neuropathological examination of MS tissue indicates that degeneration, CNS atrophy, and myelin loss are most prominent in the progressive stage, when lymphocytic inflammation paradoxically wanes. Finally, emerging clinical observations such as "progression independent of relapse activity" and "silent progression," now thought to take hold very early in the course, together argue that an underlying "cytodegenerative" process, likely targeting the myelinating unit, may in fact represent the most proximal step in a complex pathophysiological cascade exacerbated by an autoimmune inflammatory overlay. Parallels are drawn with more traditional neurodegenerative disorders, where a progressive proteopathy with prion-like propagation of toxic misfolded species is now known to play a key role. A potentially pivotal contribution of the Epstein-Barr virus and B cells in this process is also discussed.

5.
J Exp Med ; 203(2): 425-35, 2006 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-16476770

RESUMEN

The proteinase-activated receptors (PARs) are widely recognized for their modulatory properties of inflammation and neurodegeneration. We investigated the role of PAR2 in the pathogenesis of multiple sclerosis (MS) in humans and experimental autoimmune encephalomyelitis (EAE) in mice. PAR2 expression was increased on astrocytes and infiltrating macrophages in human MS and murine EAE central nervous system (CNS) white matter (P < 0.05). Macrophages and astrocytes from PAR2 wild-type (WT) and knockout (KO) mice exhibited differential immune gene expression with PAR2 KO macrophages showing significantly higher interleukin 10 production after lipopolysaccharide stimulation (P < 0.001). PAR2 activation in macrophages resulted in the release of soluble oligodendrocyte cytotoxins (P < 0.01). Myelin oligodendrocyte glycoprotein-induced EAE caused more severe inflammatory gene expression in the CNS of PAR2 WT animals (P < 0.05), together with enhanced T cell proliferation and interferon gamma production (P < 0.05), compared with KO littermates. Indeed, PAR2 WT animals showed markedly greater microglial activation and T lymphocyte infiltration accompanied by worsened demyelination and axonal injury in the CNS compared with their PAR2 KO littermates. Enhanced neuropathological changes were associated with a more severe progressive relapsing disease phenotype (P < 0.001) in WT animals. These findings reveal previously unreported pathogenic interactions between CNS PAR2 expression and neuroinflammation with ensuing demyelination and axonal injury.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Receptor PAR-2/fisiología , Animales , Astrocitos/metabolismo , Astrocitos/patología , Proliferación Celular , Células Cultivadas , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Lóbulo Frontal/metabolismo , Lóbulo Frontal/patología , Regulación de la Expresión Génica/inmunología , Humanos , Inflamación/genética , Inflamación/metabolismo , Interferón gamma/biosíntesis , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Esclerosis Múltiple/inmunología , Oligodendroglía/metabolismo , Oligodendroglía/patología , Receptor PAR-2/deficiencia , Receptor PAR-2/genética , Linfocitos T/metabolismo , Linfocitos T/patología
6.
Mol Brain ; 15(1): 67, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35883145

RESUMEN

As in neurons, CNS myelin expresses N-Methyl-D-Aspartate Receptors (NMDARs) that subserve physiological roles, but have the potential to induce injury to this vital element. Using 2-photon imaging of myelinic Ca in live ex vivo mouse optic nerves, we show that Cu ions potently modulate Ca levels in an NMDAR-dependent manner. Chelating Cu in the perfusate induced a substantial increase in Ca levels, and also caused significant axo-myelinic injury. Myelinic NMDARs are shown to be regulated by cellular prion protein; only in prion protein KO optic nerves does application of NMDA + D-serine induce a large Ca increase, consistent with strong desensitization of these receptors in the presence of prion protein limiting Ca overload. Aß1-42 peptide induced a large Ca increase that was also Cu-dependent, and was blocked by NMDAR antagonism. Our results indicate that like in neurons, myelinic NMDARs permeate potentially injurious amounts of Ca, and are also potently regulated by micromolar Cu and activated by Aß1-42 peptides. These findings shed mechanistic light on the important primary white matter injury frequently observed in Alzheimer's brain.


Asunto(s)
Vaina de Mielina , Receptores de N-Metil-D-Aspartato , Péptidos beta-Amiloides , Animales , Sistema Nervioso Central/metabolismo , Cobre/farmacología , Iones/metabolismo , Ratones , Vaina de Mielina/metabolismo , Fragmentos de Péptidos , Proteínas Priónicas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
7.
J Alzheimers Dis ; 85(4): 1721-1734, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34958041

RESUMEN

BACKGROUND: Toxic amyloid-ß (Aß) peptides aggregate into higher molecular weight assemblies and accumulate not only in the extracellular space, but also in the walls of blood vessels in the brain, increasing their permeability, and promoting immune cell migration and activation. Given the prominent role of the immune system, phagocytic blood cells may contact pathological brain materials. OBJECTIVE: To develop a novel method for early Alzheimer's disease (AD) detection, we used blood leukocytes, that could act as "sentinels" after trafficking through the brain microvasculature, to detect pathological amyloid by labelling with a conformationally-sensitive fluorescent amyloid probe and imaging with confocal spectral microscopy. METHODS: Formalin-fixed peripheral blood mononuclear cells (PBMCs) from cognitively healthy control (HC) subjects, mild cognitive impairment (MCI) and AD patients were stained with the fluorescent amyloid probe K114, and imaged. Results were validated against cerebrospinal fluid (CSF) biomarkers and clinical diagnosis. RESULTS: K114-labeled leukocytes exhibited distinctive fluorescent spectral signatures in MCI/AD subjects. Comparing subjects with single CSF biomarker-positive AD/MCI to negative controls, our technique yielded modest AUCs, which improved to the 0.90 range when only MCI subjects were included in order to measure performance in an early disease state. Combining CSF Aß42 and t-Tau metrics further improved the AUC to 0.93. CONCLUSION: Our method holds promise for sensitive detection of AD-related protein misfolding in circulating leukocytes, particularly in the early stages of disease.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico , Péptidos beta-Amiloides/metabolismo , Diagnóstico Precoz , Colorantes Fluorescentes/metabolismo , Leucocitos Mononucleares/metabolismo , Fragmentos de Péptidos/metabolismo , Enfermedad de Alzheimer/sangre , Biomarcadores/líquido cefalorraquídeo , Encéfalo/patología , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteínas tau/metabolismo
8.
J Neurosci ; 30(23): 7975-83, 2010 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-20534845

RESUMEN

A single postnatal exposure to the bacterial endotoxin, lipopolysaccharide (LPS), reduces the neuroimmune response to a subsequent LPS exposure in the adult rat. The attenuated fever and proinflammatory response is caused by a paradoxical, amplified, early corticosterone response to LPS. Here we identify the mechanisms underlying the heightened corticosterone response to LPS in adults after early life exposure to LPS. In postnatal LPS-treated rats, hypothalamic corticotrophin-releasing hormone mRNA, pituitary proopiomelanocortin mRNA, and circulating adrenocorticotrophic hormone were all increased after adult exposure to LPS without significant modification to hippocampal or hypothalamic glucocorticoid receptor mRNA or protein or vagally mediated afferent signaling to the brain. Postnatal LPS administration did cause a persistent upregulation of the LPS Toll-like receptor-4 (TLR4) mRNA in liver and spleen, but not in brain, pituitary, or adrenal gland. In addition, cyclooxygenase-2 (COX-2), which is a prostaglandin biosynthetic enzyme and is normally undetectable in most peripheral tissue, was constitutively expressed in the liver. Adult immune activation of the upregulated TLR4 and COX-2 caused a rapid, amplified rise in circulating, but not brain, prostaglandin E(2) that induced an early, enhanced activation of the hypothalamic-pituitary-adrenal (HPA) axis. Thus, postnatal LPS reprograms the neuroimmune axis by priming peripheral tissues to create a novel, prostaglandin-mediated activation of the HPA axis brought about by increased constitutive expression of TLR4 and COX-2.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Sistema Hipotálamo-Hipofisario/inmunología , Inflamación/metabolismo , Vías Nerviosas/inmunología , Sistema Hipófiso-Suprarrenal/inmunología , Receptor Toll-Like 4/metabolismo , Glándulas Suprarrenales/inmunología , Glándulas Suprarrenales/metabolismo , Hormona Adrenocorticotrópica/sangre , Animales , Western Blotting , Encéfalo/inmunología , Encéfalo/metabolismo , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/metabolismo , Ciclooxigenasa 2/genética , Hipocampo/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Hipotálamo/metabolismo , Inflamación/inducido químicamente , Inyecciones Intraperitoneales , Lipopolisacáridos , Hígado/inmunología , Hígado/metabolismo , Hipófisis/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Prostaglandinas E/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/inmunología , Bazo/metabolismo , Factores de Tiempo , Receptor Toll-Like 4/genética , Regulación hacia Arriba
9.
J Immunol ; 183(1): 298-309, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19542441

RESUMEN

The rising incidence of autoimmune diseases such as multiple sclerosis (MS) in developed countries might be due to a more hygienic environment, particularly during early life. To investigate this concept, we developed a model of neonatal exposure to a common pathogen-associated molecular pattern, LPS, and determined its impact on experimental autoimmune encephalomyelitis (EAE). Mice exposed to LPS at 2 wk of age showed a delayed onset and diminished severity of myelin oligodendrocyte glycoprotein (MOG)-induced EAE, induced at 12 wk, compared with vehicle-exposed animals. Spinal cord transcript levels of CD3epsilon and F4/80 were lower in LPS- compared with PBS-exposed EAE animals with increased IL-10 levels in the LPS-exposed group. Splenic CD11c(+) cells from LPS-exposed animals exhibited reduced MHC class II and CD83 expression but increased levels of CD80 and CD86 both before and during EAE. MOG-treated APC from LPS-exposed animals stimulated less T lymphocyte proliferation but increased expansion of CD4(+)FoxP3(+) T cells compared with APC from PBS-exposed animals. Neuropathological studies disclosed reduced myelin and axonal loss in spinal cords from LPS-exposed compared with PBS-exposed animals with EAE, and this neuroprotective effect was associated with an increased number of CD3(+)FoxP3(+) immunoreactive cells. Analyses of human brain tissue revealed that FoxP3 expression was detected in lymphocytes, albeit reduced in MS compared with non-MS patients' brains. These findings support the concept of early-life microbial exposure influencing the generation of neuroprotective regulatory T cells and may provide insights into new immunotherapeutic strategies for MS.


Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/prevención & control , Tolerancia Inmunológica , Lipopolisacáridos/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Linfocitos T Reguladores/inmunología , Animales , Animales Recién Nacidos , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Diferenciación Celular/genética , Movimiento Celular/genética , Movimiento Celular/inmunología , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/metabolismo , Células Dendríticas/patología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Factores de Transcripción Forkhead/antagonistas & inhibidores , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Tolerancia Inmunológica/genética , Interleucina-10/biosíntesis , Interleucina-10/genética , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Índice de Severidad de la Enfermedad , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología
10.
Am J Pathol ; 173(4): 1029-41, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18815152

RESUMEN

Although the physiological roles of the cellular prion protein (PrP C) remain to be fully elucidated, PrP C has been proposed to represent a potential regulator of cellular immunity. To test this hypothesis, we evaluated the consequences of PrP C deficiency on the course of experimental autoimmune encephalomyelitis induced by immunization with myelin oligodendrocyte glycoprotein peptide. Consistent with augmented proliferative responses and increased cytokine gene expression by myelin oligodendrocyte glycoprotein-primed Prnp-/- T cells, PrP C-deficient mice demonstrated more aggressive disease onset and a lack of clinical improvement during the chronic phase of experimental autoimmune encephalomyelitis. Acutely, Prnp-/- spinal cord, cerebellum, and forebrain exhibited higher levels of leukocytic infiltrates and pro-inflammatory cytokine gene expression, as well as increased spinal cord myelin basic protein and axonal loss. During the chronic phase, a remarkable persistence of leukocytic infiltrates was present in the forebrain and cerebellum, accompanied by an increase in interferon-gamma and interleukin-17 transcripts. Attenuation of T cell-dependent neuroinflammation thus represents a potential novel function of PrP C.


Asunto(s)
Encefalomielitis Autoinmune Experimental/patología , Sistema Nervioso/patología , Proteínas PrPC/deficiencia , Animales , Conducta Animal , Linfocitos T CD4-Positivos/metabolismo , Cerebelo/patología , Reactividad Cruzada , Células Dendríticas/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Inmunización , Inflamación , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas de la Mielina , Glicoproteína Asociada a Mielina , Glicoproteína Mielina-Oligodendrócito , Sistema Nervioso/metabolismo , Proteínas PrPC/metabolismo , Prosencéfalo/patología , Médula Espinal/patología , Regulación hacia Arriba
11.
FASEB J ; 22(3): 786-96, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17965263

RESUMEN

Desensitization of seven transmembrane receptors (7TMRs), which are modulated by the beta-arrestins, leads to altered G protein activation. The A1 adenosine receptor (A1AR) is an antiinflammatory 7TMR exhibiting reduced expression and activity in both multiple sclerosis (MS) and the murine MS model, experimental autoimmune encephalomyelitis (EAE) in monocytoid cells. Herein, we report that beta-arrestin-1 expression was increased in brains of MS patients relative to non-MS brains, whereas A1AR expression was concomitantly reduced. This inverse relationship between beta-arrestin-1 and A1AR was confirmed in cultured monocytoid cells as beta-arrestin-1 overexpression resulted in a down-regulation of A1AR together with the internalization of the surface receptor. Moreover, a physical interaction between beta-arrestin-1 and A1AR was demonstrated in monocytoid cells. Proinflammatory cytokines regulated the A1AR/beta-arrestin-1 interactions, while A1AR activation also modulated proinflammatory cytokines expression. During EAE, beta-arrestin-1 and A1AR expression in the spinal cord displayed a similar pattern compared to that observed in MS brains. EAE-induced neuroinflammation and neurobehavioral deficits were suppressed by glucocorticoid treatments, accompanied by concurrent reduced beta-arrestin-1 and enhanced A1AR expression. Thus, the interplay between beta-arrestin-1 and A1AR in the central nervous system during neuroinflammation represents a reciprocal regulatory mechanism through which neuroprotective therapeutic strategies for neuroinflammatory diseases might be further developed.


Asunto(s)
Arrestinas/metabolismo , Glucocorticoides/farmacología , Inmunidad Innata/efectos de los fármacos , Monocitos/inmunología , Esclerosis Múltiple/inmunología , Receptor de Adenosina A1/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo , Encefalomielitis Autoinmune Experimental/inmunología , Glucocorticoides/metabolismo , Humanos , Ratones , Esclerosis Múltiple/patología , Receptor de Adenosina A1/efectos de los fármacos , beta-Arrestina 1 , beta-Arrestinas
12.
F1000Res ; 82019.
Artículo en Inglés | MEDLINE | ID: mdl-31885862

RESUMEN

Emerging data point to important contributions of both autoimmune inflammation and progressive degeneration in the pathophysiology of multiple sclerosis (MS). Unfortunately, after decades of intensive investigation, the fundamental cause remains unknown. A large body of research on the immunobiology of MS has resulted in a variety of anti-inflammatory therapies that are highly effective at reducing brain inflammation and clinical/radiological relapses. However, despite potent suppression of inflammation, benefit in the more important and disabling progressive phase is extremely limited; thus, progressive MS has emerged as the greatest challenge for the MS research and clinical communities. Data obtained over the years point to a complex interplay between environment (e.g., the near-absolute requirement of Epstein-Barr virus exposure), immunogenetics (strong associations with a large number of immune genes), and an ever more convincing role of an underlying degenerative process resulting in demyelination (in both white and grey matter regions), axonal and neuro-synaptic injury, and a persistent innate inflammatory response with a seemingly diminishing role of T cell-mediated autoimmunity as the disease progresses. Together, these observations point toward a primary degenerative process, one whose cause remains unknown but one that entrains a nearly ubiquitous secondary autoimmune response, as a likely sequence of events underpinning this disease. Here, we briefly review what is known about the potential pathophysiological mechanisms, focus on progressive MS, and discuss the two main hypotheses of MS pathogenesis that are the topic of vigorous debate in the field: whether primary autoimmunity or degeneration lies at the foundation. Unravelling this controversy will be critically important for developing effective new therapies for the most disabling later phases of this disease.


Asunto(s)
Enfermedades Desmielinizantes , Encefalitis , Esclerosis Múltiple , Axones , Sustancia Gris , Humanos
13.
J Cell Biol ; 216(4): 1163-1181, 2017 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-28264914

RESUMEN

Cellular injury and death are ubiquitous features of disease, yet tools to detect them are limited and insensitive to subtle pathological changes. Acridine orange (AO), a nucleic acid dye with unique spectral properties, enables real-time measurement of RNA and DNA as proxies for cell viability during exposure to various noxious stimuli. This tool illuminates spectral signatures unique to various modes of cell death, such as cells undergoing apoptosis versus necrosis/necroptosis. This new approach also shows that cellular RNA decreases during necrotic, necroptotic, and apoptotic cell death caused by demyelinating, ischemic, and traumatic injuries, implying its involvement in a wide spectrum of tissue pathologies. Furthermore, cells with pathologically low levels of cytoplasmic RNA are detected earlier and in higher numbers than with standard markers including TdT-mediated dUTP biotin nick-end labeling and cleaved caspase 3 immunofluorescence. Our technique highlights AO-labeled cytoplasmic RNA as an important early marker of cellular injury and a sensitive indicator of various modes of cell death in a range of experimental models.


Asunto(s)
Naranja de Acridina/metabolismo , Apoptosis/fisiología , Muerte Celular/fisiología , Necrosis/patología , Ácidos Nucleicos/metabolismo , Animales , Caspasa 3/metabolismo , Línea Celular , Nucleótidos de Desoxiuracil/metabolismo , Humanos , Etiquetado Corte-Fin in Situ/métodos , Ratones Endogámicos C57BL , Necrosis/metabolismo , ARN/metabolismo
14.
PLoS One ; 12(11): e0188218, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29176903

RESUMEN

As an extension of the brain, the spinal cord has unique properties which could allow us to gain a better understanding of CNS pathology. The brain and cord share the same cellular components, yet the latter is simpler in cytoarchitecture and connectivity. In Alzheimer's research, virtually all focus is on brain pathology, however it has been shown that transgenic Alzheimer's mouse models accumulate beta amyloid plaques in spinal cord, suggesting that the cord possesses the same molecular machinery and conditions for plaque formation. Here we report a spatial-temporal map of plaque load in 5xFAD mouse spinal cord. We found that plaques started to appear at 11 weeks, then exhibited a time dependent increase and differential distribution along the cord. More plaques were found in cervical than other spinal levels at all time points examined. Despite heavy plaque load at 6 months, the number of cervical motor neurons in 5xFAD mice is comparable to wild type littermates. On detailed microscopic examination, fine beta amyloid-containing and beta sheet-rich thread-like structures were found in the peri-axonal space of many axons. Importantly, these novel structures appear before any plaque deposits are visible in young mice spinal cord and they co-localize with axonal swellings at later stages, suggesting that these thread-like structures might represent the initial stages of plaque formation, and could play a role in axonal damage. Additionally, we were able to demonstrate increasing myelinopathy in aged 5xFAD mouse spinal cord using the lipid probe Nile Red with high resolution. Collectively, we found significant amyloid pathology in grey and white matter of the 5xFAD mouse spinal cord which indicates that this structure maybe a useful platform to study mechanisms of Alzheimer's pathology and disease progression.


Asunto(s)
Enfermedad de Alzheimer/patología , Axones/patología , Vaina de Mielina/patología , Médula Espinal/patología , Envejecimiento , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Sustancia Gris/patología , Humanos , Ratones Transgénicos , Neuronas Motoras/patología , Neuroglía/patología , Placa Amiloide/patología , Sustancia Blanca/patología
15.
Chem Sci ; 8(8): 5636-5643, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28989601

RESUMEN

Alzheimer's disease (AD) is a multifactorial disease that is characterized by the formation of intracellular neurofibrillary tangles and extracellular amyloid-ß (Aß) plaque deposits. Increased oxidative stress, metal ion dysregulation, and the formation of toxic Aß peptide oligomers are all considered to contribute to the etiology of AD. In this work we have developed a series of ligands that are multi-target-directed in order to address several disease properties. 2-(1-(3-Hydroxypropyl)-1H-1,2,3-triazol-4-yl)phenol (POH), 2-(1-(2-morpholinoethyl)-1H-1,2,3-triazol-4-yl)phenol (PMorph), and 2-(1-(2-thiomorpholinoethyl)-1H-1,2,3-triazol-4-yl)phenol (PTMorph) have been synthesized and screened for their antioxidant capacity, Cu-binding affinity, interaction with the Aß peptide and modulation of Aß peptide aggregation, and the ability to limit Aß1-42-induced neurotoxicity in human neuronal culture. The synthetic protocol and structural variance incorporated via click chemistry, highlights the influence of R-group modification on ligand-Aß interactions and neuroprotective effects. Overall, this study demonstrates that the phenol-triazole ligand scaffold can target multiple factors associated with AD, thus warranting further therapeutic development.

16.
J Neurosci ; 24(6): 1521-9, 2004 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-14960625

RESUMEN

The neuromodulator adenosine regulates immune activation and neuronal survival through specific G-protein-coupled receptors expressed on macrophages and neurons, including the A1 adenosine receptor (A1AR). Here we show that A1AR null (A1AR-/-) mice developed a severe progressive-relapsing form of experimental allergic encephalomyelitis (EAE) compared with their wild-type (A1AR+/+) littermates. Worsened demyelination, axonal injury, and enhanced activation of microglia/macrophages were observed in A1AR-/- animals. In addition, spinal cords from A1AR-/- mice demonstrated increased proinflammatory gene expression during EAE, whereas anti-inflammatory genes were suppressed compared with A1AR+/+ animals. Macrophages from A1AR-/- animals exhibited increased expression of the proinflammatory genes, interleukin-1beta, and matrix metalloproteinase-12 on immune activation when matched with A1AR+/+ control cells. A1AR-/- macrophage-derived soluble factors caused significant oligodendrocyte cytotoxicity compared with wild-type controls. The A1AR was downregulated in microglia in A1AR+/+ mice during EAE accompanied by neuroinflammation, which recapitulated findings in multiple sclerosis (MS) patients. Caffeine treatment augmented A1AR expression on microglia, with ensuing reduction of EAE severity, which was further enhanced by concomitant treatment with the A1AR agonist, adenosine amine congener. Thus, modulation of neuroinflammation by the A1AR represents a novel mechanism that provides new therapeutic opportunities for MS and other demyelinating diseases.


Asunto(s)
Adenosina/análogos & derivados , Encefalomielitis Autoinmune Experimental/fisiopatología , Inflamación/metabolismo , Esclerosis Múltiple/fisiopatología , Vaina de Mielina/metabolismo , Receptor de Adenosina A1/metabolismo , Regulación hacia Arriba/fisiología , Adenosina/farmacología , Agonistas del Receptor de Adenosina A1 , Animales , Cafeína/farmacología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Inflamación/inmunología , Inflamación/patología , Mediadores de Inflamación/metabolismo , Interleucina-1/genética , Interleucina-1/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Metaloproteinasa 12 de la Matriz , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Ratones , Ratones Noqueados , Microglía/inmunología , Microglía/metabolismo , Microglía/patología , Esclerosis Múltiple/patología , Vaina de Mielina/inmunología , Vaina de Mielina/patología , Fenotipo , ARN Mensajero/metabolismo , Receptor de Adenosina A1/genética , Recurrencia , Índice de Severidad de la Enfermedad , Médula Espinal/patología
18.
Macromol Biosci ; 15(11): 1523-34, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26175127

RESUMEN

Dendrimers and dendriplexes, highly branched synthetic macromolecules, have gained popularity as new tools for a variety of nanomedicine strategies due to their unique structure and properties. We show that fluorescent phosphorus dendrimers are well retained by bone marrow-derived macrophages and exhibit robust spectral shift in its emission in response to polarization conditions. Fluorescence properties of this marker can also assist in identifying macrophage presence and phenotype status at different time points after spinal cord injury. Potential use of a single dendrimer compound as a drug/siRNA carrier and phenotype-specific cell tracer offers new avenues for enhanced cell therapies combined with monitoring of cell fate and function in spinal cord injury.


Asunto(s)
Células de la Médula Ósea , Rastreo Celular/métodos , Dendrímeros/farmacología , Macrófagos , Imagen Óptica/métodos , Traumatismos de la Médula Espinal , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Células Cultivadas , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Nanomedicina/métodos , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología
19.
Exp Anim ; 52(1): 43-52, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12638236

RESUMEN

Studies were conducted in C57BL/6N Crj male mice and in cultured hepatocytes to clarify the relationship between galactosamine (GaIN) induced apoptosis and [Ca2+]i kinetics. Chlorpromazine (CPZ), a Ca(2+)-calmodulin antagonist, and verapamil (VR), a Ca(2+)-channel blocker each inhibited GaIN-induced DNA fragmentation and the appearance of apoptotic bodies. The kinetics of calcium uptake were evaluated using a calcium analyzer with the acetoxymethyl ester of fura-PE3 (fura-PE3/AM, 2.5 microM) as the calcium reporter. An increase in [Ca2+]i was detected in the cultured hepatocytes within 3 hours after treatment with 20 mM GaIN; this increase was inhibited by pretreatment with either 20 microM CPZ or 30 microM VR. Ca2+ imaging by confocal laser scanning microscopy showed that increase in [Ca2+]i after treatment with GaIN was initially localized around nuclei, while [Ca2+]i signals were later diffuse and observed throughout the cytoplasm. The activities of lactate dehydrogenase (LDH) and serum glutamate-pyruvate transaminase (sGPT), used as indicators of plasma membrane damage and leakage, however, were not reduced by pretreatment with CPZ or VR. From these findings, we infer that the DNA fragmentation in GaIN-induced hepatocyte apoptosis is associated with an elevation in the perinuclear concentration of Ca2+, but GaIN-induced necrotic cell death is triggered through pathway(s) that are insensitive to blockage of Ca2+ influx and therefore appear to occur independently of elevation in [Ca2+]i. These results help to clarify the role of calcium flux in hepatocyte apoptosis and necrosis induced by exposure to hepatotoxins in vivo and in vitro.


Asunto(s)
Apoptosis/efectos de los fármacos , Bloqueadores de los Canales de Calcio/farmacología , Calmodulina/antagonistas & inhibidores , Clorpromazina/farmacología , Galactosamina/antagonistas & inhibidores , Galactosamina/toxicidad , Hepatocitos/citología , Hepatocitos/metabolismo , Verapamilo/farmacología , Animales , Apoptosis/genética , Calcio/metabolismo , Señalización del Calcio/fisiología , Células Cultivadas , Fragmentación del ADN/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL
20.
Front Cell Dev Biol ; 2: 45, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25364752

RESUMEN

Although it is well established that misfolding of the cellular prion protein (PrP(C)) into the ß-sheet-rich, aggregated scrapie conformation (PrP(Sc)) causes a variety of transmissible spongiform encephalopathies (TSEs), the physiological roles of PrP(C) are still incompletely understood. There is accumulating evidence describing the roles of PrP(C) in neurodegeneration and neuroinflammation. Recently, we identified a functional regulation of NMDA receptors by PrP(C) that involves formation of a physical protein complex between these proteins. Excessive NMDA receptor activity during conditions such as ischemia mediates enhanced Ca(2+) entry into cells and contributes to excitotoxic neuronal death. In addition, NMDA receptors and/or PrP(C) play critical roles in neuroinflammation and glial cell toxicity. Inhibition of NMDA receptor activity protects against PrP(Sc)-induced neuronal death. Moreover, in mice lacking PrP(C), infarct size is increased after focal cerebral ischemia, and absence of PrP(C) increases susceptibility of neurons to NMDA receptor-dependent death. Recently, PrP(C) was found to be a receptor for oligomeric beta-amyloid (Aß) peptides, suggesting a role for PrP(C) in Alzheimer's disease (AD). Our recent findings suggest that Aß peptides enhance NMDA receptor current by perturbing the normal copper- and PrP(C)-dependent regulation of these receptors. Here, we review evidence highlighting a role for PrP(C) in preventing NMDA receptor-mediated excitotoxicity and inflammation. There is a need for more detailed molecular characterization of PrP(C)-mediated regulation of NMDA receptors, such as determining which NMDA receptor subunits mediate pathogenic effects upon loss of PrP(C)-mediated regulation and identifying PrP(C) binding site(s) on the receptor. This knowledge will allow development of novel therapeutic interventions for not only TSEs, but also for AD and other neurodegenerative disorders involving dysfunction of PrP(C).

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA