Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Neurobiol Dis ; 76: 46-56, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25662335

RESUMEN

Huntington disease (HD) is an inherited, fatal neurodegenerative disease with no disease-modifying therapy currently available. In addition to characteristic motor deficits and atrophy of the caudate nucleus, signature hallmarks of HD include behavioral abnormalities, immune activation, and cortical and white matter loss. The identification and validation of novel therapeutic targets that contribute to these degenerative cellular processes may lead to new interventions that slow or even halt the course of this insidious disease. Semaphorin 4D (SEMA4D) is a transmembrane signaling molecule that modulates a variety of processes central to neuroinflammation and neurodegeneration including glial cell activation, neuronal growth cone collapse and apoptosis of neural precursors, as well as inhibition of oligodendrocyte migration, differentiation and process formation. Therefore, inhibition of SEMA4D signaling could reduce CNS inflammation, increase neuronal outgrowth and enhance oligodendrocyte maturation, which may be of therapeutic benefit in the treatment of several neurodegenerative diseases, including HD. To that end, we evaluated the preclinical therapeutic efficacy of an anti-SEMA4D monoclonal antibody, which prevents the interaction between SEMA4D and its receptors, in the YAC128 transgenic HD mouse model. Anti-SEMA4D treatment ameliorated neuropathological signatures, including striatal atrophy, cortical atrophy, and corpus callosum atrophy and prevented testicular degeneration in YAC128 mice. In parallel, a subset of behavioral symptoms was improved in anti-SEMA4D treated YAC128 mice, including reduced anxiety-like behavior and rescue of cognitive deficits. There was, however, no discernible effect on motor deficits. The preservation of brain gray and white matter and improvement in behavioral measures in YAC128 mice treated with anti-SEMA4D suggest that this approach could represent a viable therapeutic strategy for the treatment of HD. Importantly, this work provides in vivo demonstration that inhibition of pathways initiated by SEMA4D constitutes a novel approach to moderation of neurodegeneration.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos CD/inmunología , Enfermedad de Huntington/terapia , Semaforinas/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Encéfalo/metabolismo , Encéfalo/patología , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/terapia , Modelos Animales de Enfermedad , Enfermedad de Huntington/complicaciones , Inmunoterapia , Ratones , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
2.
Neurobiol Dis ; 73: 254-68, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25461192

RESUMEN

Multiple sclerosis (MS) is a chronic neuroinflammatory disease characterized by immune cell infiltration of CNS, blood-brain barrier (BBB) breakdown, localized myelin destruction, and progressive neuronal degeneration. There exists a significant need to identify novel therapeutic targets and strategies that effectively and safely disrupt and even reverse disease pathophysiology. Signaling cascades initiated by semaphorin 4D (SEMA4D) induce glial activation, neuronal process collapse, inhibit migration and differentiation of oligodendrocyte precursor cells (OPCs), and disrupt endothelial tight junctions forming the BBB. To target SEMA4D, we generated a monoclonal antibody that recognizes mouse, rat, monkey and human SEMA4D with high affinity and blocks interaction between SEMA4D and its cognate receptors. In vitro, anti-SEMA4D reverses the inhibitory effects of recombinant SEMA4D on OPC survival and differentiation. In vivo, anti-SEMA4D significantly attenuates experimental autoimmune encephalomyelitis in multiple rodent models by preserving BBB integrity and axonal myelination and can be shown to promote migration of OPC to the site of lesions and improve myelin status following chemically-induced demyelination. Our study underscores SEMA4D as a key factor in CNS disease and supports the further development of antibody-based inhibition of SEMA4D as a novel therapeutic strategy for MS and other neurologic diseases with evidence of demyelination and/or compromise to the neurovascular unit.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Oligodendroglía/metabolismo , Semaforinas/metabolismo , Animales , Anticuerpos Monoclonales , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Semaforinas/antagonistas & inhibidores , Semaforinas/inmunología
3.
Am J Pathol ; 182(6): 2285-97, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23567638

RESUMEN

Neuroinflammation, through production of proinflammatory molecules and activated glial cells, is implicated in Alzheimer's disease (AD) pathogenesis. One such proinflammatory mediator is tumor necrosis factor α (TNF-α), a multifunctional cytokine produced in excess and associated with amyloid ß-driven inflammation and cognitive decline. Long-term global inhibition of TNF receptor type I (TNF-RI) and TNF-RII signaling without cell or stage specificity in triple-transgenic AD mice exacerbates hallmark amyloid and neurofibrillary tangle pathology. These observations revealed that long-term pan anti-TNF-α inhibition accelerates disease, cautions against long-term use of anti-TNF-α therapeutics for AD, and urges more selective regulation of TNF signaling. We used adeno-associated virus vector-delivered siRNAs to selectively knock down neuronal TNF-R signaling. We demonstrate divergent roles for neuronal TNF-RI and TNF-RII where loss of opposing TNF-RII leads to TNF-RI-mediated exacerbation of amyloid ß and Tau pathology in aged triple-transgenic AD mice. Dampening of TNF-RII or TNF-RI+RII leads to a stage-independent increase in Iba-1-positive microglial staining, implying that neuronal TNF-RII may act nonautonomously on the microglial cell population. These results reveal that TNF-R signaling is complex, and it is unlikely that all cells and both receptors will respond positively to broad anti-TNF-α treatments at various stages of disease. In aggregate, these data further support the development of cell-, stage-, and/or receptor-specific anti-TNF-α therapeutics for AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Neuronas/metabolismo , Receptores del Factor de Necrosis Tumoral/biosíntesis , Adenoviridae/genética , Envejecimiento/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/patología , Progresión de la Enfermedad , Regulación hacia Abajo/fisiología , Técnicas de Silenciamiento del Gen , Vectores Genéticos , Masculino , Ratones , Ratones Transgénicos , Microglía/metabolismo , Placa Amiloide/metabolismo , ARN Interferente Pequeño/genética , Receptores del Factor de Necrosis Tumoral/deficiencia , Receptores del Factor de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Receptores Tipo II del Factor de Necrosis Tumoral/deficiencia , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Transducción de Señal/fisiología
4.
Proc Natl Acad Sci U S A ; 108(2): 852-7, 2011 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-21187387

RESUMEN

Target-derived neurotrophins use retrogradely transported Trk-signaling endosomes to promote survival and neuronal phenotype at the soma. Despite their critical role in neurotrophin signaling, the nature and molecular composition of these endosomes remain largely unknown, the result of an inability to specifically identify the retrograde signaling entity. Using EGF-bound nanoparticles and chimeric, EGF-binding TrkB receptors, we elucidate Trk-endosomal events involving their formation, processing, retrograde transport, and somal signaling in sympathetic neurons. By comparing retrograde endosomal signaling by Trk to the related but poorly neuromodulatory EGF-receptor, we find that Trk and EGF-receptor endosomes are formed and processed by distinct mechanisms. Surprisingly, Trk and EGF-receptors are both retrogradely transported to the soma in multivesicular bodies. However, only the Trk-multivesicular bodies rely on Pincher-dependent macroendocytosis and processing. Retrograde signaling through Pincher-generated Trk-multivesicular bodies is distinctively refractory to signal termination by lysosomal processing, resulting in sustained somal signaling and neuronal gene expression.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Endosomas/metabolismo , Proteínas del Tejido Nervioso/fisiología , Proteínas Nucleares/fisiología , Receptor trkA/metabolismo , Transducción de Señal , Animales , Endocitosis , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Microscopía Fluorescente/métodos , Células PC12 , Ratas , Proteínas de Unión al GTP rab5/metabolismo
5.
Cureus ; 16(6): e62224, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-39006590

RESUMEN

Colonic xanthomas are a rare finding, particularly when combined with a tubular adenoma in a single polyp. While transformation to malignancy is not thought to be higher than that of a tubular adenoma alone, there is still concern as to the pathophysiology of xanthoma formation within the colon and what that may mean for patient outcomes. Here, we present a patient undergoing a routine screening colonoscopy with the removal of a rectosigmoid polyp consistent with xanthoma and tubular adenoma histopathology. Proper follow-up for identification of possible metabolic derangements and increased colonic surveillance is recommended to mitigate the risk of further xanthoma or adenocarcinoma formation.

6.
Hum Mol Genet ; 20(R1): R28-41, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21429918

RESUMEN

Genetic therapy is undergoing a renaissance with expansion of viral and synthetic vectors, use of oligonucleotides (RNA and DNA) and sequence-targeted regulatory molecules, as well as genetically modified cells, including induced pluripotent stem cells from the patients themselves. Several clinical trials for neurologic syndromes appear quite promising. This review covers genetic strategies to ameliorate neurologic syndromes of different etiologies, including lysosomal storage diseases, Alzheimer's disease and other amyloidopathies, Parkinson's disease, spinal muscular atrophy, amyotrophic lateral sclerosis and brain tumors. This field has been propelled by genetic technologies, including identifying disease genes and disruptive mutations, design of genomic interacting elements to regulate transcription and splicing of specific precursor mRNAs and use of novel non-coding regulatory RNAs. These versatile new tools for manipulation of genetic elements provide the ability to tailor the mode of genetic intervention to specific aspects of a disease state.


Asunto(s)
Terapia Genética/métodos , Enfermedades del Sistema Nervioso/terapia , Enfermedades Neurodegenerativas/terapia , Enfermedad de Alzheimer/terapia , Vectores Genéticos/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Enfermedades por Almacenamiento Lisosomal/terapia , Enfermedad de Parkinson/terapia , Empalme del ARN
7.
PLoS Genet ; 6(4): e1000902, 2010 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-20386743

RESUMEN

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are associated with late-onset, autosomal-dominant, familial Parkinson's disease (PD) and also contribute to sporadic disease. The LRRK2 gene encodes a large protein with multiple domains, including functional Roc GTPase and protein kinase domains. Mutations in LRRK2 most likely cause disease through a toxic gain-of-function mechanism. The expression of human LRRK2 variants in cultured primary neurons induces toxicity that is dependent on intact GTP binding or kinase activities. However, the mechanism(s) underlying LRRK2-induced neuronal toxicity is poorly understood, and the contribution of GTPase and/or kinase activity to LRRK2 pathobiology is not well defined. To explore the pathobiology of LRRK2, we have developed a model of LRRK2 cytotoxicity in the baker's yeast Saccharomyces cerevisiae. Protein domain analysis in this model reveals that expression of GTPase domain-containing fragments of human LRRK2 are toxic. LRRK2 toxicity in yeast can be modulated by altering GTPase activity and is closely associated with defects in endocytic vesicular trafficking and autophagy. These truncated LRRK2 variants induce similar toxicity in both yeast and primary neuronal models and cause similar vesicular defects in yeast as full-length LRRK2 causes in primary neurons. The toxicity induced by truncated LRRK2 variants in yeast acts through a mechanism distinct from toxicity induced by human alpha-synuclein. A genome-wide genetic screen identified modifiers of LRRK2-induced toxicity in yeast including components of vesicular trafficking pathways, which can also modulate the trafficking defects caused by expression of truncated LRRK2 variants. Our results provide insight into the basic pathobiology of LRRK2 and suggest that the GTPase domain may contribute to the toxicity of LRRK2. These findings may guide future therapeutic strategies aimed at attenuating LRRK2-mediated neurodegeneration.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , GTP Fosfohidrolasas/genética , Genoma Fúngico , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Mutación , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
8.
J Neurosci ; 31(49): 17788-99, 2011 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-22159095

RESUMEN

Elevating spinal levels of neurotrophin NT-3 (NT3) while increasing expression of the NR2D subunit of the NMDA receptor using a HSV viral construct promotes formation of novel multisynaptic projections from lateral white matter (LWM) axons to motoneurons in neonates. However, this treatment is ineffective after postnatal day 10. Because chondroitinase ABC (ChABC) treatment restores plasticity in the adult CNS, we have added ChABC to this treatment and applied the combination to adult rats receiving a left lateral hemisection (Hx) at T8. All hemisected animals initially dragged the ipsilateral hindpaw and displayed abnormal gait. Rats treated with ChABC or NT3/HSV-NR2D recovered partial hindlimb locomotor function, but animals receiving combined therapy displayed the most improved body stability and interlimb coordination [Basso-Beattie-Bresnahan (BBB) locomotor scale and gait analysis]. Electrical stimulation of the left LWM at T6 did not evoke any synaptic response in ipsilateral L5 motoneurons of control hemisected animals, indicating interruption of the white matter. Only animals with the full combination treatment recovered consistent multisynaptic responses in these motoneurons indicating formation of a detour pathway around the Hx. These physiological findings were supported by the observation of increased branching of both cut and intact LWM axons into the gray matter near the injury. ChABC-treated animals displayed more sprouting than control animals and those receiving NT3/HSV-NR2D; animals receiving the combination of all three treatments showed the most sprouting. Our results indicate that therapies aimed at increasing plasticity, promoting axon growth and modulating synaptic function have synergistic effects and promote better functional recovery than if applied individually.


Asunto(s)
Axones/metabolismo , Condroitina ABC Liasa/metabolismo , Plasticidad Neuronal/fisiología , Neurotrofina 3/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/fisiopatología , Análisis de Varianza , Animales , Axones/patología , Biotina/análogos & derivados , Biotina/metabolismo , Células Cultivadas , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Dextranos/metabolismo , Modelos Animales de Enfermedad , Potenciales Postsinápticos Excitadores , Femenino , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Hiperalgesia/fisiopatología , Locomoción/fisiología , Ratas , Ratas Sprague-Dawley , Transfección , beta-Galactosidasa/metabolismo
9.
Am J Pathol ; 179(4): 2053-70, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21835156

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by severe memory loss and cognitive impairment. Neuroinflammation, including the extensive production of pro-inflammatory molecules and the activation of microglia, has been implicated in the disease process. Tumor necrosis factor (TNF)-α, a prototypic pro-inflammatory cytokine, is elevated in AD, is neurotoxic, and colocalizes with amyloid plaques in AD animal models and human brains. We previously demonstrated that the expression of TNF-α is increased in AD mice at ages preceding the development of hallmark amyloid and tau pathological features and that long-term expression of this cytokine in these mice leads to marked neuronal death. Such observations suggest that TNF-α signaling promotes AD pathogenesis and that therapeutics suppressing this cytokine's activity may be beneficial. To dissect TNF-α receptor signaling requirements in AD, we generated triple-transgenic AD mice (3xTg-AD) lacking both TNF-α receptor 1 (TNF-RI) and 2 (TNF-RII), 3xTg-ADxTNF-RI/RII knock out, the cognate receptors of TNF-α. These mice exhibit enhanced amyloid and tau-related pathological features by the age of 15 months, in stark contrast to age-matched 3xTg-AD counterparts. Moreover, 3xTg-ADxTNF-RI/RII knock out-derived primary microglia reveal reduced amyloid-ß phagocytic marker expression and phagocytosis activity, indicating that intact TNF-α receptor signaling is critical for microglial-mediated uptake of extracellular amyloid-ß peptide pools. Overall, our results demonstrate that globally ablated TNF receptor signaling exacerbates pathogenesis and argues against long-term use of pan-anti-TNF-α inhibitors for the treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Encéfalo/metabolismo , Encéfalo/patología , Receptores Tipo II del Factor de Necrosis Tumoral/deficiencia , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Envejecimiento/patología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/fisiopatología , Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Región CA1 Hipocampal/patología , Región CA1 Hipocampal/fisiopatología , Región CA3 Hipocampal/patología , Región CA3 Hipocampal/fisiopatología , Proteínas de Unión al Calcio/metabolismo , Cruzamientos Genéticos , Femenino , Humanos , Receptores de Lipopolisacáridos/metabolismo , Potenciación a Largo Plazo , Masculino , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Microglía/metabolismo , Microglía/patología , Fagocitosis , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Sinapsis/metabolismo , Transgenes/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas tau/genética
10.
J Pediatr Orthop ; 32(5): 473-6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22706462

RESUMEN

INTRODUCTION: Fractures of the distal third of the radius in children are common and most heal without incident. However, distal radial shaft malunion with apex volar angulation may lead to volar dislocation of the distal ulna with forearm supination, although it has been rarely reported. The aim of this study was to document 2 such cases and to make recommendations regarding the management of these patients. METHODS: We report the cases of 2 boys, ages 6 and 8 years, who sustained radial shaft fractures that healed with apex volar angulation and who developed intractable volar dislocation of the distal ulna in adolescence. In both cases, corrective radial osteotomy at the site of the malunion restored full stability of the distal radial-ulnar joint without the need for soft-tissue reconstruction or ulnar styloid nonunion repair. DISCUSSION: This injury pattern is rarely reported but should be considered in cases of repeated volar dislocation of the distal ulna with supination. We recommend a corrective osteotomy at the site of the malunion to restore stability to the distal radial-ulnar joint. LEVEL OF EVIDENCE: IV.


Asunto(s)
Osteotomía/métodos , Fracturas del Radio/patología , Cúbito/patología , Adolescente , Fracturas Mal Unidas , Humanos , Luxaciones Articulares , Inestabilidad de la Articulación , Masculino , Fracturas del Radio/cirugía , Supinación
11.
J Neurosci ; 30(28): 9567-77, 2010 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-20631185

RESUMEN

Clinical studies to date have failed to establish therapeutic benefit of glial cell-derived neurotrophic factor (GDNF) in Parkinson's disease (PD). In contrast to previous nonclinical neuroprotective reports, this study shows clinically relevant and long-lasting regeneration of the dopaminergic system in rhesus macaques lesioned with 1-methy-4-phenyl-1,2,3,6-tetrahydropyridine 3-6 months before GDNF gene delivery (AAV2-GDNF). The observed progressive amelioration of functional deficits, recovery of dopamine, and regrowth of fibers to the striatal neuropil demonstrate that high GDNF expression in the putamen promotes restoration of the dopaminergic system in a primate model of advanced PD. Extensive distribution of GDNF within the putamen and transport to the severely lesioned substantia nigra, after convection-enhanced delivery of AAV2-GDNF into the putamen, indicates anterograde transport via striatonigral connections and is anticipated to occur in PD patients. Overall, these data demonstrate nonclinical neurorestoration after putaminal infusion of AAV2-GDNF and suggest that clinical investigation in PD patients is warranted.


Asunto(s)
Dopamina/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/uso terapéutico , Regeneración Nerviosa/genética , Neuronas/metabolismo , Trastornos Parkinsonianos/terapia , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Análisis de Varianza , Animales , Encéfalo/metabolismo , Encéfalo/patología , Cromatografía Líquida de Alta Presión , Ensayo de Inmunoadsorción Enzimática , Femenino , Terapia Genética , Vectores Genéticos , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Inmunohistoquímica , Macaca mulatta , Masculino , Neuronas/patología , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Recuperación de la Función
12.
Glia ; 59(4): 627-40, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21294162

RESUMEN

White matter pathology has been documented in the brains of familial Alzheimer's disease (FAD)-afflicted individuals during presymptomatic and preclinical stages of AD. How these defects in myelination integrity arise and what roles they may play in AD pathophysiology have yet to be fully elucidated. We previously demonstrated that triple-transgenic AD (3xTg-AD) mice, which harbor the human amyloid precursor Swedish mutation, presenilin-1 M146V (PS1(M146V) ) knock-in mutation, and tau(P301L) mutation, exhibit myelin abnormalities analogous to FAD patients and that Aß(1-42) contributes to these white matter deficits. Herein, we demonstrate that the PS1(M146V) mutation predisposes mouse oligodendrocyte precursor (mOP) cells to Aß(1-42) -induced alterations in cell differentiation in vitro. Furthermore, PS1(M146V) expression compromised mOP cell function and MBP protein distribution, a process that is further aggravated with exposure to Aß(1-42) . We found that the myelination defect and MBP subcellular mislocalization triggered by PS1(M146V) and Aß(1-42) can be effectively prevented by treatment with the GSK-3ß inhibitor, TWS119, thereby implicating GSK-3ß kinase activity in this pathogenic cascade. Overall, this work provides further mechanistic insights into PS1(M146V) and Aß(1-42) -driven oligodendrocyte dysfunction andmyelin damage during early presymptomatic stages of AD, and provides a new target in oligodendrocytes for developing therapies designed to avert AD-related white matter pathology.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Diferenciación Celular/fisiología , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Presenilina-1/genética , Péptidos beta-Amiloides/farmacología , Análisis de Varianza , Animales , Western Blotting , Diferenciación Celular/efectos de los fármacos , Línea Celular , Citometría de Flujo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Inmunohistoquímica , Ratones , Ratones Transgénicos , Mutación , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/genética , Vaina de Mielina/patología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/patología , Presenilina-1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Eur J Neurosci ; 34(8): 1256-67, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21995852

RESUMEN

To encourage re-establishment of functional innervation of ipsilateral lumbar motoneurons by descending fibers after an intervening lateral thoracic (T10) hemisection (Hx), we treated adult rats with the following agents: (i) anti-Nogo-A antibodies to neutralize the growth-inhibitor Nogo-A; (ii) neurotrophin-3 (NT-3) via engineered fibroblasts to promote neuron survival and plasticity; and (iii) the NMDA-receptor 2d (NR2d) subunit via an HSV-1 amplicon vector to elevate NMDA receptor function by reversing the Mg(2+) block, thereby enhancing synaptic plasticity and promoting the effects of NT-3. Synaptic responses evoked by stimulation of the ventrolateral funiculus ipsilateral and rostral to the Hx were recorded intracellularly from ipsilateral lumbar motoneurons. In uninjured adult rats short-latency (1.7-ms) monosynaptic responses were observed. After Hx these monosynaptic responses were abolished. In the Nogo-Ab + NT-3 + NR2d group, long-latency (approximately 10 ms), probably polysynaptic, responses were recorded and these were not abolished by re-transection of the spinal cord through the Hx area. This suggests that these novel responses resulted from new connections established around the Hx. Anterograde anatomical tracing from the cervical grey matter ipsilateral to the Hx revealed increased numbers of axons re-crossing the midline below the lesion in the Nogo-Ab + NT-3 + NR2d group. The combined treatment resulted in slightly better motor function in the absence of adverse effects (e.g. pain). Together, these results suggest that the combination treatment with Nogo-Ab + NT-3 + NR2d can produce a functional 'detour' around the lesion in a laterally hemisected spinal cord. This novel combination treatment may help to improve function of the damaged spinal cord.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Proteínas de la Mielina/inmunología , Neurotrofina 3/farmacología , Subunidades de Proteína/farmacología , Receptores de N-Metil-D-Aspartato/uso terapéutico , Traumatismos de la Médula Espinal/patología , Médula Espinal/efectos de los fármacos , Animales , Conducta Animal/fisiología , Femenino , Humanos , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Neuronas Motoras/citología , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/fisiología , Neurotrofina 3/uso terapéutico , Proteínas Nogo , Subunidades de Proteína/uso terapéutico , Desempeño Psicomotor , Ratas , Ratas Sprague-Dawley , Médula Espinal/patología , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/fisiopatología
14.
Am J Pathol ; 177(3): 1422-35, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20696774

RESUMEN

The detection of myelin disruptions in Alzheimer's disease (AD)-affected brain raises the possibility that oligodendrocytes undergo pathophysiological assault over the protracted course of this neurodegenerative disease. Oligodendrocyte compromise arising from direct toxic effects imparted by pathological amyloid-beta peptides and/or through signals derived from degenerating neurons could play an important role in the disease process. We previously demonstrated that 3xTg-AD mice, which harbor the human amyloid precursor protein Swedish mutant transgene, presenilin knock-in mutation, and tau P301L mutant transgene, exhibit significant alterations in overall myelination patterns and oligodendrocyte status at time points preceding the appearance of amyloid and tau pathology. Herein, we demonstrate that Abeta(1-42) leads to increased caspase-3 expression and apoptotic cell death of both nondifferentiated and differentiated mouse oligodendrocyte precursor (mOP) cells in vitro. Through use of a recombinant adeno-associated virus serotype-2 (rAAV2) vector expressing an Abeta(1-42)-specific intracellular antibody (intrabody), oligodendrocyte and myelin marker expression, as well as myelin integrity, were restored in the vector-infused brain regions of 3xTg-AD mice. Overall, this work provides further insights into the impact of Abeta(1-42)-mediated toxicity on the temporal and spatial progression of subtle myelin disruption during the early presymptomatic stages of AD and may help to validate new therapeutic options designed to avert these early impairments.


Asunto(s)
Corteza Entorrinal/patología , Vaina de Mielina/patología , Oligodendroglía/patología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Análisis de Varianza , Animales , Apoptosis/genética , Western Blotting , Caspasa 3/genética , Caspasa 3/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Corteza Entorrinal/metabolismo , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , Microscopía Electrónica , Vaina de Mielina/genética , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
15.
Blood ; 113(14): 3264-75, 2009 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-18952891

RESUMEN

Pattern recognition receptors represent the first line of defense against invading pathogens. Herpes simplex virus (HSV) encodes multiple ligands detected by these receptors, yet persists in the majority of infected individuals indicating a breakdown in host defense against the virus. Here we identify a novel mechanism through which HSV immediate-early protein ICP0 inhibits TLR-dependent inflammatory response by blocking NF-kappaB and JNK activation downstream of TLR signal activation. This process depends on ICP0-mediated translocation of USP7 (HAUSP) from the nucleus to cytoplasm. We show that nuclear USP7 migrates to the cytoplasm in response to TLR engagement, a process that contributes to termination of TLR response. Cytoplasmic USP7 binds to and deubiquitinates TRAF6 and IKKgamma, thus terminating TLR-mediated NF-kappaB and JNK activation. These findings suggest that USP7 is part of a negative feedback loop regulating TLR signaling and that ICP0 exploits this physiologic process to attenuate innate response to HSV. ICP0 inhibition of the TLR response serves to uncouple the innate and adaptive immune response, thereby playing a key role in HSV pathogenesis and persistence.


Asunto(s)
Proteínas Inmediatas-Precoces/fisiología , Inmunidad Innata/fisiología , Receptores Toll-Like/fisiología , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina-Proteína Ligasas/fisiología , Células Cultivadas , Herpes Simple/genética , Herpes Simple/inmunología , Humanos , Quinasa I-kappa B/metabolismo , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Inmunidad Innata/genética , FN-kappa B/metabolismo , FN-kappa B/fisiología , Unión Proteica/genética , Unión Proteica/fisiología , Procesamiento Proteico-Postraduccional/genética , Transporte de Proteínas , Factor 6 Asociado a Receptor de TNF/metabolismo , Ubiquitina/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Peptidasa Específica de Ubiquitina 7
17.
Nat Med ; 9(2): 231-5, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12539041

RESUMEN

Antigen-specific T cells demonstrate several potent effector functions during immune responses. Direct killing of infected cells is crucial for clearing viruses and other intracellular pathogens, but it has been difficult to measure the frequency of cytolytic cells. We have now developed a single-cell assay to measure the number of cytotoxic cells in a population, using a herpes simplex virus amplicon vector to express Escherichia coli beta-galactosidase in mouse or human target cells, and an Elispot to detect release of beta-galactosidase from killed target cells. This antigen-specific, perforin-dependent Lysispot assay has been combined with a cytokine Elispot in a two-color assay to confirm that cytotoxicity and interferon-gamma secretion are regulated independently. The simultaneous enumeration of cytokine-secreting and cytotoxic cells should be invaluable for ex vivo analysis of immune responses during infection and autoimmunity.


Asunto(s)
Interferón gamma/metabolismo , Linfocitos T Citotóxicos/citología , Animales , Escherichia coli/enzimología , Vectores Genéticos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Simplexvirus/genética , Linfocitos T Citotóxicos/inmunología , beta-Galactosidasa/genética
18.
Mol Ther ; 18(8): 1471-81, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20551911

RESUMEN

Alzheimer's disease (AD) is a progressive dementing disorder characterized by age-related amyloid-beta (Abeta) deposition, neurofibrillary tangles, and synapse and neuronal loss. It is widely recognized that Abeta is a principal pathogenic mediator of AD. Our goal was to develop an immunotherapeutic approach, which would specifically lead to the clearance and/or neutralization of Abeta in the triple transgenic mouse model (3xTg-AD). These mice develop the amyloid and tangle pathologies and synaptic dysfunction reminiscent of human AD. Using a human single-chain variable fragment (scFv) antibody phage display library, a novel scFv antibody specific to Abeta was isolated, its activity characterized in vitro, and its open reading frame subsequently cloned into a recombinant adeno-associated virus (rAAV) vector. Three-month-old 3xTg-AD mice were intrahippocampally infused with serotype-1 rAAV vectors encoding Abeta-scFv or a control vector using convection-enhanced delivery (CED). Mice receiving rAAV1-Abeta-scFv harbored lower levels of insoluble Abeta and hyperphosphorylated tau, and exhibited improved cognitive function as measured by the Morris Water Maze (MWM) spatial memory task. These results underscore the potential of gene-based passive vaccination for AD, and provide further rationale for the development of Abeta-targeting strategies for this debilitating disease.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/inmunología , Dependovirus/genética , Aprendizaje por Laberinto/fisiología , Anticuerpos de Cadena Única/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Células Cultivadas , Cricetinae , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Anticuerpos de Cadena Única/genética
19.
J Biol Chem ; 284(40): 27557-66, 2009 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-19666470

RESUMEN

Tumor necrosis factor-alpha (TNF-alpha), a proinflammatory cytokine, has been implicated as a central mediator in multiple homeostatic and pathologic processes. Signaling cascades downstream of its cellular cognate receptors, as well as the resultant transcriptional responses have received intense interest in regards to how such signals impact cellular physiology. Notably, TNF-alpha was shown to potentiate neuronal Ca(2+) signaling by enhancing type-1 inositol 1,4,5-trisphosphate receptor (IP(3)R) steady-state mRNA levels. In the present study, we sought to determine the promoter region ultimately responsive to TNF-alpha exposure. We report that a sequence encompassing a specificity protein 1 (SP-1) binding site is necessary for TNF-alpha regulation. Electrophoretic mobility shift analysis demonstrated specific binding to this sequence, while site-directed mutagenesis of this site abrogated both JNK-mediated regulation as well as transcription factor binding. Expression of a dominant-negative SP-1 eliminated both the enhanced promoter activity and the elevated IP(3)R-mediated Ca(2+) signals observed with TNF-alpha exposure. Overall, these data delineate a key pathway by which TNF-alpha in a neuronal environment modulates IP(3)R expression and intracellular Ca(2+) homeostasis.


Asunto(s)
Regulación de la Expresión Génica , Receptores de Inositol 1,4,5-Trifosfato/genética , Regiones Promotoras Genéticas/genética , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Calcio/metabolismo , Línea Celular Tumoral , Ensayo de Cambio de Movilidad Electroforética , Humanos , Inflamación/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Datos de Secuencia Molecular , Mutación , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Factor de Transcripción Sp1/metabolismo , Activación Transcripcional
20.
Am J Pathol ; 175(5): 2076-88, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19808651

RESUMEN

Inflammatory processes, including the episodic and/ or chronic elaboration of cytokines, have been identified as playing key roles in a number of neurological disorders. Whether these activities impart a disease-resolving and/or contributory outcome depends at least in part on the disease context, stage of pathogenesis, and cellular milieu in which these factors are released. Interferon-gamma (IFNgamma) is one such cytokine that produces pleiotropic effects in the brain. It is protective by ensuring maintenance of virus latency after infection, yet deleterious by recruiting and activating microglia that secrete potentially damaging factors at sites of brain injury. Using the triple-transgenic mouse model of Alzheimer's disease (3xTg-AD), which develops amyloid and tau pathologies in a pattern reminiscent of human Alzheimer's disease, we initiated chronic intrahippocampal expression of IFNgamma through delivery of a serotype-1 recombinant adeno-associated virus vector (rAAV1-IFNgamma). Ten months of IFNgamma expression led to an increase in microglial activation, steady-state levels of proinflammatory cytokine and chemokine transcripts, and severity of amyloid-related pathology. In contrast, these rAAV1-IFNgamma-treated 3xTg-AD mice also exhibited diminished phospho-tau pathology and evidence of increased neurogenesis. Overall, IFNgamma mediates what seem to be diametrically opposed functions in the setting of AD-related neurodegeneration. Gaining an understanding as to how these apparently divergent functions are interrelated and controlled could elucidate new therapeutic strategies designed to harness the neuroprotective activity of IFNgamma.


Asunto(s)
Enfermedad de Alzheimer , Interferón gamma/inmunología , Ratones Transgénicos , Neurogénesis/fisiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Biomarcadores/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Interferón gamma/genética , Ratones , Microglía/citología , Microglía/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA