Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Immunity ; 35(3): 349-60, 2011 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-21835646

RESUMEN

The transport of calcium ions (Ca(2+)) to the cytosol is essential for immunoreceptor signaling, regulating lymphocyte differentiation, activation, and effector function. Increases in cytosolic-free Ca(2+) concentrations are thought to be mediated through two interconnected and complementary mechanisms: the release of endoplasmic reticulum Ca(2+) "stores" and "store-operated" Ca(2+) entry via plasma membrane channels. However, the identity of molecular components conducting Ca(2+) currents within developing and mature T cells is unclear. Here, we have demonstrated that the L-type "voltage-dependent" Ca(2+) channel Ca(V)1.4 plays a cell-intrinsic role in the function, development, and survival of naive T cells. Plasma membrane Ca(V)1.4 was found to be essential for modulation of intracellular Ca(2+) stores and T cell receptor (TCR)-induced rises in cytosolic-free Ca(2+), impacting activation of Ras-extracellular signal-regulated kinase (ERK) and nuclear factor of activated T cells (NFAT) pathways. Collectively, these studies revealed that Ca(V)1.4 functions in controlling naive T cell homeostasis and antigen-driven T cell immune responses.


Asunto(s)
Canales de Calcio/genética , Canales de Calcio/metabolismo , Homeostasis , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Animales , Calcio/metabolismo , Canales de Calcio/deficiencia , Canales de Calcio Tipo L , Proliferación Celular , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción NFATC/metabolismo , Linfocitos T/citología , Proteínas ras/metabolismo
2.
Brain Commun ; 4(2): fcac006, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35474853

RESUMEN

Cardiorespiratory arrest and death in mouse models of sudden unexpected death in epilepsy occur when spreading depolarization is triggered by cortical seizures and then propagates to the brainstem. However, the critical brain regions and the specific changes required to allow spreading depolarization to propagate to the brainstem under the relatively rare circumstances leading to a fatal seizure are unknown. We previously found that following cortical seizure-inducing electrical stimulation, spreading depolarization could occur in both the superior and inferior colliculi in Cacna1aS218L mice, but was never observed in wild-type animals or following non-seizure-inducing stimuli in Cacna1aS218L mice. Here, we show that optogenetic stimulation of the superior/inferior colliculi in Cacna1aS218L mice induces severe seizures, and resulting spreading depolarization in the superior/inferior colliculi that propagates to the brainstem and correlates with the respiratory arrest followed by cardiac arrest. Further, we show that neurons of the superior colliculus in Cacna1aS218L mice exhibit hyperexcitable properties that we propose underlie a distinct susceptibility to spreading depolarization. Our data suggest that the susceptibility of the superior colliculus to elicit fatal spreading depolarization is a result of either genetic or seizure-related alterations within the superior colliculus that may involve changes to structure, connectivity and/or excitability.

3.
J Neurotrauma ; 35(24): 2861-2871, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30113266

RESUMEN

Spinal cord injury (SCI) is a devastating neurological condition for which there is no effective treatment to restore neurological function. The development of new treatments for those with SCI may be hampered by the insensitivity of clinical tools to assess motor function in humans. Treatments aimed at preserving neuronal function through anti-inflammatory pathways (i.e., neuroprotection) have been a mainstay of pre-clinical SCI research for decades. Minocycline, a clinically available antibiotic agent with anti-inflammatory properties, has demonstrated promising neuroprotective effects in a variety of animal models and improved motor recovery in a Phase-2 human trial. Here, we leveraged our recently developed T3 severe contusion model in the rat to determine the ability of minocycline to preserve descending sympathoexcitatory axons and improve cardiovascular control after SCI. Forty-one male Wistar rats were randomized to either a treatment group (minocycline; n = 20) or a control group (vehicle; n = 21). All rats received a severe T3 contusion. Minocycline (or vehicle) was administered intraperitoneally at one hour post-injury (90 mg/kg), then every 12 h for two weeks (45 mg/kg). Neuroanatomical correlates (lesion area, descending sympathoexcitatory axons) were assessed, in addition to an assessment of cardiovascular control (hemodynamics, autonomic dysreflexia) and motor behavior. Here, we show that minocycline reduces lesion area, increases the number of descending sympathoexctitatory axons traversing the injury site, and ultimately reduces the severity of autonomic dysreflexia. Finally, we show that autonomic dysreflexia is a more sensitive marker of treatment stratification than motor function.


Asunto(s)
Disreflexia Autónoma/etiología , Minociclina/farmacología , Fármacos Neuroprotectores/farmacología , Traumatismos de la Médula Espinal/complicaciones , Médula Espinal/efectos de los fármacos , Animales , Antiinflamatorios/farmacología , Disreflexia Autónoma/patología , Disreflexia Autónoma/fisiopatología , Modelos Animales de Enfermedad , Masculino , Distribución Aleatoria , Ratas , Ratas Wistar , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología
4.
Sci Rep ; 3: 1354, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23446889

RESUMEN

Pathogenic neoangiogenesis in Alzheimer's disease (AD) is due to amyloid-beta (Aß) and results in blood-brain barrier (BBB) leakiness in AD. It likely occurs as a compensatory response to impaired cerebral blood flow and provides a strong link between brain vascularity and AD. Aß immunotherapy is an experimental treatment for AD; however, unexpected negative vascular side effects seen in early human clinical trials demonstrate that our knowledge of Aß and AD pathogenesis is incomplete. We demonstrate that immunization with Aß peptides neutralizes the amyloid trigger leading to neoangiogenesis and reverses hypervascularity in Tg2576 AD mice. This process resolves plaque burden suggesting that neoangiogenesis is a key mechanism underlying plaque formation. A meta-analysis demonstrated that hypervascular reversion in vaccinated Alzheimer's patients. This appears to be the first example of vascular reversion following any therapeutic intervention and supports the conclusion that modulation of neoangiogenesis may repair damage in the AD brain.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Vacunas contra el Alzheimer/inmunología , Péptidos beta-Amiloides/inmunología , Neovascularización Patológica/inmunología , Enfermedad de Alzheimer/tratamiento farmacológico , Vacunas contra el Alzheimer/administración & dosificación , Péptidos beta-Amiloides/administración & dosificación , Animales , Barrera Hematoencefálica/patología , Encéfalo/irrigación sanguínea , Modelos Animales de Enfermedad , Femenino , Inmunización , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Patológica/tratamiento farmacológico , Uniones Estrechas/patología
5.
PLoS One ; 6(8): e23789, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21909359

RESUMEN

Evidence of reduced blood-brain barrier (BBB) integrity preceding other Alzheimer's disease (AD) pathology provides a strong link between cerebrovascular angiopathy and AD. However, the "Vascular hypothesis", holds that BBB leakiness in AD is likely due to hypoxia and neuroinflammation leading to vascular deterioration and apoptosis. We propose an alternative hypothesis: amyloidogenesis promotes extensive neoangiogenesis leading to increased vascular permeability and subsequent hypervascularization in AD. Cerebrovascular integrity was characterized in Tg2576 AD model mice that overexpress the human amyloid precursor protein (APP) containing the double missense mutations, APPsw, found in a Swedish family, that causes early-onset AD. The expression of tight junction (TJ) proteins, occludin and ZO-1, were examined in conjunction with markers of apoptosis and angiogenesis. In aged Tg2576 AD mice, a significant increase in the incidence of disrupted TJs, compared to age matched wild-type littermates and young mice of both genotypes, was directly linked to an increased microvascular density but not apoptosis, which strongly supports amyloidogenic triggered hypervascularity as the basis for BBB disruption. Hypervascularity in human patients was corroborated in a comparison of postmortem brain tissues from AD and controls. Our results demonstrate that amylodogenesis mediates BBB disruption and leakiness through promoting neoangiogenesis and hypervascularity, resulting in the redistribution of TJs that maintain the barrier and thus, provides a new paradigm for integrating vascular remodeling with the pathophysiology observed in AD. Thus the extensive angiogenesis identified in AD brain, exhibits parallels to the neovascularity evident in the pathophysiology of other diseases such as age-related macular degeneration.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Amiloide/metabolismo , Barrera Hematoencefálica/fisiopatología , Encéfalo/irrigación sanguínea , Encéfalo/patología , Neovascularización Patológica/patología , Envejecimiento/patología , Animales , Apoptosis , Barrera Hematoencefálica/patología , Encéfalo/fisiopatología , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Microvasos/patología , Microvasos/fisiopatología , Neovascularización Patológica/complicaciones , Neovascularización Patológica/fisiopatología , Permeabilidad , Transducción de Señal , Uniones Estrechas/metabolismo , Uniones Estrechas/patología
6.
Cancer Res ; 68(23): 9601-7, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19047136

RESUMEN

Histone deacetylase inhibitors (HDACi) have been hailed as a powerful new class of anticancer drugs. The HDACi, trichostatin A (TSA), is thought to interfere with epigenetic control of cell cycle progression in G1 and G2-M phase, resulting in growth arrest, differentiation, or apoptosis. Here, we describe a novel mechanism of action of HDACis in promoting immune responses against tumors. We report that treatment of carcinoma cells with TSA increases the expression of many components of the antigen processing machinery, including TAP-1, TAP-2, LMP-2, and Tapasin. Consistent with this result, we found that treatment of metastatic carcinoma cells with TSA also results in an increase in MHC class I expression on the cell surface that functionally translates into an enhanced susceptibility to killing by antigen-specific CTLs. Finally, we observed that TSA treatment suppresses tumor growth and increases tap-1 promoter activity in TAP-deficient tumor cells in vivo. Intriguingly, this in vivo anti-tumoral effect of TSA is entirely mediated by an increase in immunogenicity of the tumor cells, as it does not occur in immunodeficient mice. These novel insights into the molecular mechanisms controlling tumor immune escape may help revise immunotherapeutic modalities for eradicating cancers.


Asunto(s)
Antígenos de Neoplasias/inmunología , Inhibidores Enzimáticos/farmacología , Inhibidores de Histona Desacetilasas , Ácidos Hidroxámicos/farmacología , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/inmunología , Transportador de Casetes de Unión a ATP, Subfamilia B, Miembro 2 , Miembro 3 de la Subfamilia B de Transportadores de Casetes de Unión a ATP , Transportadoras de Casetes de Unión a ATP/biosíntesis , Transportadoras de Casetes de Unión a ATP/genética , Animales , Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/genética , Antígenos de Neoplasias/biosíntesis , Línea Celular Tumoral , Epigénesis Genética/inmunología , Antígenos de Histocompatibilidad Clase I/biosíntesis , Antígenos de Histocompatibilidad Clase I/inmunología , Melanoma Experimental/genética , Proteínas de Transporte de Membrana/biosíntesis , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , ARN Polimerasa II/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología
7.
Mol Cell Biol ; 27(22): 7886-94, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17875943

RESUMEN

Downregulation of the transporter associated with antigen processing 1 (TAP-1) has been observed in many tumors and is closely associated with tumor immunoevasion mechanisms, growth, and metastatic ability. The molecular mechanisms underlying the relatively low level of transcription of the tap-1 gene in cancer cells are largely unexplained. In this study, we tested the hypothesis that epigenetic regulation plays a fundamental role in controlling tumor antigen processing and immune escape mechanisms. We found that the lack of TAP-1 transcription in TAP-deficient cells correlated with low levels of recruitment of the histone acetyltransferase, CBP, to the TAP-1 promoter. This results in lower levels of histone H3 acetylation at the TAP-1 promoter, leading to a decrease in accessibility of the RNA polymerase II complex to the TAP-1 promoter. These observations suggest that CBP-mediated histone H3 acetylation normally relaxes the chromatin structure around the TAP-1 promoter region, allowing transcription. In addition, we found a hitherto-unknown mechanism wherein interferon gamma up-regulates TAP-1 expression by increasing histone H3 acetylation at the TAP-1 promoter locus. These findings lie at the heart of understanding immune escape mechanisms in tumors and suggest that the reversal of epigenetic codes may provide novel immunotherapeutic paradigms for intervention in cancer.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Carcinoma , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Invasividad Neoplásica , Metástasis de la Neoplasia , Transportador de Casetes de Unión a ATP, Subfamilia B, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Acetilación , Animales , Secuencia de Bases , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Carcinoma/genética , Carcinoma/inmunología , Carcinoma/patología , Línea Celular , Genes MHC Clase I , Histonas/metabolismo , Humanos , Interferón gamma/metabolismo , Complejo Mayor de Histocompatibilidad , Ratones , Datos de Secuencia Molecular , Invasividad Neoplásica/genética , Invasividad Neoplásica/inmunología , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/inmunología , Regiones Promotoras Genéticas , ARN Polimerasa II/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
8.
Vaccine ; 25(12): 2331-9, 2007 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-17229499

RESUMEN

Despite continued progress in understanding the pathophysiology of tumours, curative therapeutic options are still lacking for the metastatic form of the disease. One approach that has gathered considerable interest is the creation of therapeutic vaccines using genetically engineered non-replicating viruses as vehicles to revive immunosurveillance mechanisms that may eradicate residual tumour cells. A perceived problem with this approach is that the number of non-replicating viruses used as a vaccine inoculum does not remotely approximate the total number of cells in the body, nor even the number of tumour cells in the case of large tumour burden or metastasis. Here, we addressed the hypothesis that a limited amount of inoculum (1x10(8) PFU) of recombinant non-replicating adenovirus encoding human TAP1 (AdhTAP1) can induce protective immunity against 1.5x10(5) TAP-deficient, metastatic melanoma cells transplanted into a normal mouse (total of approximately 1x10(11) body cells). We show that efficacious anti-tumour cytolytic T cell responses are indeed induced by injecting melanoma-bearing animals with small numbers of recombinant viruses, resulting in increases in tumour-infiltrating dendritic cells, enhanced memory T cell subpopulations and, most importantly, in increased animal survival. This novel approach uses a limited input inoculum relative to the tumour cell mass, and thus achieves an efficacious outcome that has so far eluded other vaccine, immunotherapeutic or gene therapeutic strategies where there is a requisite for the majority of tumour cells to be transduced for beneficial outcome to be achieved.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/inmunología , Adenoviridae/genética , Vacunas contra el Cáncer/inmunología , Melanoma Experimental/inmunología , Linfocitos T/inmunología , Transportador de Casetes de Unión a ATP, Subfamilia B, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/genética , Línea Celular Tumoral , Células Dendríticas/inmunología , Femenino , Antígenos H-2/inmunología , Antígenos H-2/metabolismo , Humanos , Immunoblotting , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA