Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Int J Mol Sci ; 20(9)2019 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-31052389

RESUMEN

Apolipoprotein E (apoE), a key lipid transport protein in the brain, is predominantly produced by astrocytes. Astrocytes are the most numerous cell type in the brain and are the main support network for neurons. They play a critical role in the synthesis and delivery of cholesterol in the brain. Humans have three common apoE isoforms, apoE2, apoE3 and apoE4, that show a strong genotype effect on the risk and age of onset for sporadic and late onset forms of Alzheimer's disease (AD). Carriers of an ε4 allele have an increased risk of developing AD, while those with an ε2 allele are protected. Investigations into the contribution of apoE to the development of AD has yielded conflicting results and there is still much speculation about the role of this protein in disease. Here, we review the opposing hypotheses currently described in the literature and the approaches that have been considered for targeting apoE as a novel therapeutic strategy for AD. Additionally, we provide our perspective on the rationale for targeting apoE and the challenges that arise with respect to "drug-ability" of this target.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Apolipoproteínas E/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Animales , Apolipoproteínas E/química , Apolipoproteínas E/genética , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Humanos , Terapia Molecular Dirigida , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico
2.
J Neuroinflammation ; 15(1): 142, 2018 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-29759062

RESUMEN

BACKGROUND: Acute neurological insults caused by infection, systemic inflammation, ischemia, or traumatic injury are often associated with breakdown of the blood-brain barrier (BBB) followed by infiltration of peripheral immune cells, cytotoxic proteins, and water. BBB breakdown and extravasation of these peripheral components into the brain parenchyma result in inflammation, oxidative stress, edema, excitotoxicity, and neurodegeneration. These downstream consequences of BBB dysfunction can drive pathophysiological processes and play a substantial role in the morbidity and mortality of acute and chronic neurological insults, and contribute to long-term sequelae. Preserving or rescuing BBB integrity and homeostasis therefore represents a translational research area of high therapeutic potential. METHODS: Induction of general and localized BBB disruption in mice was carried out using systemic administration of LPS and focal photothrombotic ischemic insult, respectively, in the presence and absence of the monoacylglycerol lipase (MAGL) inhibitor, CPD-4645. The effects of CPD-4645 treatment were assessed by gene expression analysis performed on neurovascular-enriched brain fractions, cytokine and inflammatory mediator measurement, and functional assessment of BBB permeability. The mechanism of action of CPD-4645 was studied pharmacologically using inverse agonists/antagonists of the cannabinoid receptors CB1 and CB2. RESULTS: Here, we demonstrate that the neurovasculature exhibits a unique transcriptional signature following inflammatory insults, and pharmacological inhibition of MAGL using a newly characterized inhibitor rescues the transcriptional profile of brain vasculature and restores its functional homeostasis. This pronounced effect of MAGL inhibition on blood-brain barrier permeability is evident following both systemic inflammatory and localized ischemic insults. Mechanistically, the protective effects of the MAGL inhibitor are partially mediated by cannabinoid receptor signaling in the ischemic brain insult. CONCLUSIONS: Our results support considering MAGL inhibitors as potential therapeutics for BBB dysfunction and cerebral edema associated with inflammatory brain insults.


Asunto(s)
Ácidos Araquidónicos/antagonistas & inhibidores , Ácidos Araquidónicos/metabolismo , Barrera Hematoencefálica/metabolismo , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Permeabilidad Capilar/fisiología , Endocannabinoides/antagonistas & inhibidores , Endocannabinoides/metabolismo , Glicéridos/antagonistas & inhibidores , Glicéridos/metabolismo , Animales , Barrera Hematoencefálica/efectos de los fármacos , Lesiones Encefálicas/inducido químicamente , Permeabilidad Capilar/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Hidrólisis/efectos de los fármacos , Lipopolisacáridos , Masculino , Ratones , Ratones Endogámicos C57BL , Monoacilglicerol Lipasas/antagonistas & inhibidores , Monoacilglicerol Lipasas/metabolismo
3.
Blood ; 121(6): 1008-15, 2013 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-23243271

RESUMEN

The majority of peripheral serotonin is stored in platelets, which secrete it on activation. Serotonin releases Weibel-Palade bodies (WPBs) and we asked whether absence of platelet serotonin affects neutrophil recruitment in inflammatory responses. Tryptophan hydroxylase (Tph)1­deficient mice, lacking non-neuronal serotonin, showed mild leukocytosis compared with wild-type (WT), primarily driven by an elevated neutrophil count. Despite this, 50% fewer leukocytes rolled on unstimulated mesenteric venous endothelium of Tph1(-/-) mice. The velocity of rolling leukocytes was higher in Tph1(-/-) mice, indicating fewer selectin-mediated interactions with endothelium. Stimulation of endothelium with histamine, a secretagogue of WPBs, or injection of serotonin normalized the rolling in Tph1(-/-) mice. Diminished rolling in Tph1(-/-) mice resulted in reduced firm adhesion of leukocytes after lipopolysaccharide treatment. Blocking platelet serotonin uptake with fluoxetine in WT mice reduced serum serotonin by > 80% and similarly reduced leukocyte rolling and adhesion. Four hours after inflammatory stimulation, neutrophil extravasation into lung, peritoneum, and skin wounds was reduced in Tph1(-/-) mice, whereas in vitro neutrophil chemotaxis was independent of serotonin. Survival of lipopolysaccharide-induced endotoxic shock was improved in Tph1(-/-) mice. In conclusion, platelet serotonin promotes the recruitment of neutrophils in acute inflammation, supporting an important role for platelet serotonin in innate immunity.


Asunto(s)
Plaquetas/inmunología , Inflamación/inmunología , Neutrófilos/inmunología , Serotonina/inmunología , Enfermedad Aguda , Animales , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Quimiotaxis/efectos de los fármacos , Quimiotaxis/inmunología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Citometría de Flujo , Fluoxetina/inmunología , Fluoxetina/farmacología , Histamina/inmunología , Histamina/farmacología , Inflamación/genética , Inflamación/metabolismo , Estimación de Kaplan-Meier , Selectina L/inmunología , Selectina L/metabolismo , Rodamiento de Leucocito/efectos de los fármacos , Rodamiento de Leucocito/genética , Rodamiento de Leucocito/inmunología , Leucocitosis/genética , Leucocitosis/inmunología , Leucocitosis/metabolismo , Lipopolisacáridos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/inmunología , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Serotonina/sangre , Serotonina/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/inmunología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Choque Séptico/inducido químicamente , Choque Séptico/genética , Choque Séptico/inmunología , Triptófano Hidroxilasa/deficiencia , Triptófano Hidroxilasa/genética , Cuerpos de Weibel-Palade/efectos de los fármacos , Cuerpos de Weibel-Palade/inmunología , Cuerpos de Weibel-Palade/metabolismo
4.
Arterioscler Thromb Vasc Biol ; 33(9): 2112-20, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23825365

RESUMEN

OBJECTIVE: Aberrant blood-brain barrier (BBB) permeability is a hallmark pathology of many central nervous system diseases. von Willebrand factor (VWF) is stored in endothelial Weibel-Palade bodies from where it is released on activation into plasma and basement membrane. The role of VWF in endothelial homeostasis is unclear. The goal of this study was to assess the role of VWF in disease models associated with increased BBB permeability. APPROACH AND RESULTS: We did not find any differences in BBB permeability to Evans blue dye at baseline between wild-type and VWF(-/-) animals. We next used 2 models presenting with increased BBB permeability, hypoxia/reoxygenation and pilocarpine-induced status epilepticus, to assess the response of VWF(-/-) animals. In both models, VWF(-/-) mice maintained a tighter BBB than wild-type mice. VWF(-/-) mice fared worse in both conditions, with ≈ 100% of VWF(-/-) mice dying within 120 minutes after pilocarpine administration, whereas >80% of wild-type animals survived. Investigation into the status of tight junction proteins revealed that VWF(-/-) mice expressed more claudin-5 at baseline. In vitro work confirmed that the presence of subendothelial VWF is inhibitory to claudin-5 expression. CONCLUSIONS: VWF deficiency confers partial preservation of BBB integrity after hypoxia/reoxygenation and seizures. Surprisingly, this decrease in BBB permeability did not result in protection of animals because they demonstrated more severe pathology in both models compared with wild-type animals. These data suggest that a rigid BBB is detrimental (to the organism) during certain disease states and that VWF release may provide desired flexibility under stress.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Permeabilidad Capilar , Células Endoteliales/metabolismo , Hipoxia-Isquemia Encefálica/prevención & control , Estado Epiléptico/prevención & control , Enfermedades de von Willebrand/metabolismo , Factor de von Willebrand/metabolismo , Animales , Células Cultivadas , Claudina-5/metabolismo , Modelos Animales de Enfermedad , Femenino , Genotipo , Hipoxia-Isquemia Encefálica/genética , Hipoxia-Isquemia Encefálica/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Pilocarpina , Proteínas Recombinantes/administración & dosificación , Estado Epiléptico/inducido químicamente , Estado Epiléptico/genética , Estado Epiléptico/metabolismo , Trombocitopenia/metabolismo , Uniones Estrechas/metabolismo , Enfermedades de von Willebrand/genética , Factor de von Willebrand/administración & dosificación , Factor de von Willebrand/genética
5.
Arterioscler Thromb Vasc Biol ; 32(8): 1884-91, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22628431

RESUMEN

OBJECTIVE: Recently, a growing number of studies have revealed a prothrombotic and cytotoxic role for extracellular chromatin. Cerebral ischemia/reperfusion injury is characterized by a significant amount of cell death and neutrophil activation, both of which may result in the release of chromatin. The goal of this study was to assess the effect of extracellular chromatin in ischemic stroke using a mouse model of transient middle cerebral artery occlusion. METHODS AND RESULTS: Similar to reports in stroke patients, we observed increased levels of circulating nucleosomes and DNA after ischemic stroke in mice. In addition, we observed that general hypoxia also augmented extracellular chromatin. We hypothesized that targeting extracellular chromatin components would be protective in ischemic stroke. Indeed, treatment with recombinant human DNase 1 significantly improved stroke outcome. Neutralization of histones using an antihistone antibody was also protective as evidenced by smaller infarct volumes, whereas increasing levels of extracellular histones via histone infusion exacerbated stroke outcome by increasing infarct size and worsening functional outcome. CONCLUSIONS: Our results indicate that extracellular chromatin is generated and is detrimental during cerebral ischemia/reperfusion in mice. Targeting DNA and histones may be a new therapeutic strategy to limit injury resulting from ischemic stroke.


Asunto(s)
Isquemia Encefálica/etiología , Cromatina/fisiología , Accidente Cerebrovascular/etiología , Animales , Isquemia Encefálica/tratamiento farmacológico , Desoxirribonucleasa I/uso terapéutico , Histonas/antagonistas & inhibidores , Hipoxia/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Nucleosomas/fisiología , Accidente Cerebrovascular/tratamiento farmacológico
6.
J Neuroinflammation ; 9: 218, 2012 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-22985494

RESUMEN

BACKGROUND: Altered permeability of the blood-brain barrier (BBB) is a feature of numerous neurological conditions including multiple sclerosis, cerebral malaria, viral hemorrhagic fevers and acute hemorrhagic leukoencephalitis. Our laboratory has developed a murine model of CD8 T cell-initiated central nervous system (CNS) vascular permeability in which vascular endothelial growth factor (VEGF) signaling plays a prominent role in BBB disruption. FINDINGS: In this study, we addressed the hypothesis that in vivo blockade of VEGF signal transduction through administration of peptide (ATWLPPR) to inhibit neuropilin-1 (NRP-1) would have a therapeutic effect following induction of CD8 T cell-initiated BBB disruption. We report that inhibition of NRP-1, a co-receptor that enhances VEGFR2 (flk-1) receptor activation, decreases vascular permeability, brain hemorrhage, and mortality in this model of CD8 T cell-initiated BBB disruption. We also examine the expression pattern of VEGFR2 (flk-1) and VEGFR1 (flt-1) mRNA expression during a time course of this condition. We find that viral infection of the brain leads to increased expression of flk-1 mRNA. In addition, flk-1 and flt-1 expression levels decrease in the striatum and hippocampus in later time points following induction of CD8 T cell-mediated BBB disruption. CONCLUSION: This study demonstrates that NRP-1 is a potential therapeutic target in neuro-inflammatory diseases involving BBB disruption and brain hemorrhage. Additionally, the reduction in VEGF receptors subsequent to BBB disruption could be involved in compensatory negative feedback as an attempt to reduce vascular permeability.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Permeabilidad Capilar/fisiología , Sistema Nervioso Central/fisiología , Neuropilina-1/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Neuropilina-1/antagonistas & inhibidores , Péptidos/farmacología , ARN Mensajero/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
7.
J Neuroinflammation ; 9: 60, 2012 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-22452799

RESUMEN

BACKGROUND: The extent to which susceptibility to brain hemorrhage is derived from blood-derived factors or stromal tissue remains largely unknown. We have developed an inducible model of CD8 T cell-initiated blood-brain barrier (BBB) disruption using a variation of the Theiler's murine encephalomyelitis virus (TMEV) model of multiple sclerosis. This peptide-induced fatal syndrome (PIFS) model results in severe central nervous system (CNS) vascular permeability and death in the C57BL/6 mouse strain, but not in the 129 SvIm mouse strain, despite the two strains' having indistinguishable CD8 T-cell responses. Therefore, we hypothesize that hematopoietic factors contribute to susceptibility to brain hemorrhage, CNS vascular permeability and death following induction of PIFS. METHODS: PIFS was induced by intravenous injection of VP2121-130 peptide at 7 days post-TMEV infection. We then investigated brain inflammation, astrocyte activation, vascular permeability, functional deficit and microhemorrhage formation using T2*-weighted magnetic resonance imaging (MRI) in C57BL/6 and 129 SvIm mice. To investigate the contribution of hematopoietic cells in this model, hemorrhage-resistant 129 SvIm mice were reconstituted with C57BL/6 or autologous 129 SvIm bone marrow. Gadolinium-enhanced, T1-weighted MRI was used to visualize the extent of CNS vascular permeability after bone marrow transfer. RESULTS: C57BL/6 and 129 SvIm mice had similar inflammation in the CNS during acute infection. After administration of VP2121-130 peptide, however, C57BL/6 mice had increased astrocyte activation, CNS vascular permeability, microhemorrhage formation and functional deficits compared to 129 SvIm mice. The 129 SvIm mice reconstituted with C57BL/6 but not autologous bone marrow had increased microhemorrhage formation as measured by T2*-weighted MRI, exhibited a profound increase in CNS vascular permeability as measured by three-dimensional volumetric analysis of gadolinium-enhanced, T1-weighted MRI, and became moribund in this model system. CONCLUSION: C57BL/6 mice are highly susceptible to microhemorrhage formation, severe CNS vascular permeability and morbidity compared to the 129 SvIm mouse. This susceptibility is transferable with the bone marrow compartment, demonstrating that hematopoietic factors are responsible for the onset of brain microhemorrhage and vascular permeability in immune-mediated fatal BBB disruption.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Infecciones por Cardiovirus/complicaciones , Hemorragias Intracraneales/etiología , Animales , Astrocitos/efectos de los fármacos , Barrera Hematoencefálica , Trasplante de Médula Ósea/métodos , Linfocitos T CD8-positivos/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Proteínas de la Cápside/efectos adversos , Modelos Animales de Enfermedad , Citometría de Flujo , Fluoresceína-5-Isotiocianato/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Hematínicos , Hemorragias Intracraneales/cirugía , Hemorragias Intracraneales/virología , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos , Actividad Motora/fisiología , Prueba de Desempeño de Rotación con Aceleración Constante , Theilovirus/patogenicidad , Proteínas Virales/efectos adversos
8.
J Immunol ; 184(2): 1031-40, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20008293

RESUMEN

Dysregulation of the blood-brain barrier (BBB) is a hallmark feature of numerous neurologic disorders as diverse as multiple sclerosis, stroke, epilepsy, viral hemorrhagic fevers, cerebral malaria, and acute hemorrhagic leukoencephalitis. CD8 T cells are one immune cell type that have been implicated in promoting vascular permeability in these conditions. Our laboratory has created a murine model of CD8 T cell-mediated CNS vascular permeability using a variation of the Theiler's murine encephalomyelitis virus system traditionally used to study multiple sclerosis. Previously, we demonstrated that CD8 T cells have the capacity to initiate astrocyte activation, cerebral endothelial cell tight junction protein alterations and CNS vascular permeability through a perforin-dependent process. To address the downstream mechanism by which CD8 T cells promote BBB dysregulation, in this study, we assess the role of vascular endothelial growth factor (VEGF) expression in this model. We demonstrate that neuronal expression of VEGF is significantly upregulated prior to, and coinciding with, CNS vascular permeability. Phosphorylation of fetal liver kinase-1 is significantly increased early in this process indicating activation of this receptor. Specific inhibition of neuropilin-1 significantly reduced CNS vascular permeability and fetal liver kinase-1 activation, and preserved levels of the cerebral endothelial cell tight junction protein occludin. Our data demonstrate that CD8 T cells initiate neuronal expression of VEGF in the CNS under neuroinflammatory conditions, and that VEGF may be a viable therapeutic target in neurologic disease characterized by inflammation-induced BBB disruption.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Permeabilidad Capilar , Sistema Nervioso Central/irrigación sanguínea , Regulación de la Expresión Génica/inmunología , Inflamación , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Barrera Hematoencefálica/metabolismo , Sistema Nervioso Central/patología , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Fosforilación , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
9.
PLoS One ; 14(9): e0216263, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31487284

RESUMEN

Explants of embryonic chick sympathetic and sensory ganglia were found to exhibit asymmetric radial outgrowth of neurites under standard culture conditions with or without exogenous Nerve Growth Factor [NGF]. Opposing sides of an explant exhibited: a) differences in neurite length and, b) differences in neurite morphology. Strikingly, this asymmetry exhibited co-orientation among segregated, neighboring explants. The underlying mechanism(s) of the asymmetry and its co-orientation are not known but appear to depend on cell clustering because dissociated sympathetic neurons do not exhibit co-orientation whereas re-aggregated clusters of cells do. This emergent behavior may be similar to the community effect described in other cell types. If a similar phenomenon exists in the embryo, or in maturity, it may contribute to the establishment of proper orientation of neurite outgrowth during development and/or injury-induced neuronal plasticity.


Asunto(s)
Ganglios Sensoriales/citología , Proyección Neuronal , Cultivo Primario de Células/métodos , Técnicas de Cultivo de Tejidos/métodos , Animales , Embrión de Pollo
10.
J Neuroinflammation ; 5: 31, 2008 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-18606015

RESUMEN

Acute hemorrhagic leukoencephalomyelitis (AHLE) is a rare neurological condition characterized by the development of acute hemorrhagic demyelination and high mortality. The pathomechanism of AHLE, as well as potential therapeutic approaches, have remained elusive due to the lack of suitable animal models. We report the first murine model of AHLE using a variation of the Theiler's Murine Encephalitis Virus (TMEV) MS model. During acute TMEV infection, C57BL/6 mice do not normally undergo demyelination. However, when 7 day TMEV infected C57BL/6 mice are intravenously administered the immunodominant CD8 T cell peptide, VP2121-130, animals develop characteristics of human AHLE based on pathologic, MRI and clinical features including microhemorrhages, increased blood-brain barrier permeability, and demyelination. The animals also develop severe disability as assessed using the rotarod assay. This study demonstrates the development of hemorrhagic demyelination in TMEV infected C57BL/6 mice within 24 hours of inducing this condition through intravenous administration of CD8 T cell restricted peptide. This study is also the first demonstration of rapid demyelination in a TMEV resistant non-demyelinating strain without transgenic alterations or pharmacologically induced immunosuppression.


Asunto(s)
Enfermedades Desmielinizantes/patología , Leucoencefalitis Hemorrágica Aguda/patología , Esclerosis Múltiple/patología , Animales , Encéfalo/patología , Enfermedades Desmielinizantes/fisiopatología , Modelos Animales de Enfermedad , Leucoencefalitis Hemorrágica Aguda/fisiopatología , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/fisiopatología
11.
PLoS One ; 13(11): e0207241, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30439993

RESUMEN

Prevalence of depression is higher in patients with cancer than in the general population. Sustained systemic inflammation has been associated with depressive behavior and it has been reported that depressed patients commonly display alterations in their immune system. We previously showed that cancer in mice induces a systemic environment that promotes neutrophil activation and leukocytosis. We thus hypothesized that the peripheral systemic response to a solid tumor leads to endothelial activation, which may promote inflammatory changes in the brain with behavioral consequences. Using the Lewis lung carcinoma (LLC) model, we show that tumor growth induces a progressive increase in peripheral inflammation as observed by elevated interleukin-6 (IL-6). In behavioral studies, tumor-bearing mice showed no sign of motor, coordination or short term working memory deficits as assessed by rotarod, balance-beam, and novel object recognition tests. However, there was an impairment in the grip strength test and interestingly, an anxious and despair-like phenotype in the elevated plus-maze, and tail suspension tests, respectively. Immunostaining of perfused brains revealed fibrin accumulation in the vasculature with some leakage into the parenchyma, a process known to activate endothelial cells. Taken together, our results suggest that the inflamed and prothrombotic systemic environment created by the growth of a peripherally-located solid tumor induces endothelial activation, accumulation of fibrin in the brain and astrocyte activation, perhaps leading to depressive-like behavior.


Asunto(s)
Astrocitos/inmunología , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/psicología , Depresión/inmunología , Inflamación/metabolismo , Inflamación/psicología , Animales , Ansiedad/inmunología , Ansiedad/patología , Ansiedad/psicología , Astrocitos/patología , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/patología , Permeabilidad Capilar , Carcinoma Pulmonar de Lewis/patología , Línea Celular Tumoral , Depresión/etiología , Depresión/patología , Células Endoteliales/inmunología , Células Endoteliales/patología , Femenino , Fibrinógeno/metabolismo , Inflamación/patología , Interleucina-6/sangre , Memoria , Ratones Endogámicos C57BL , Actividad Motora
12.
Blood Adv ; 2(9): 954-963, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29700007

RESUMEN

Alzheimer disease (AD) is a neurodegenerative disorder characterized by extracellular ß-amyloid (Aß) deposition. Although peripheral inflammation and cerebrovascular pathology are reported in AD, there is a lack of plasma biomarkers in this field. Because the contact system is triggered in patient plasma, we hypothesized that the hemostasis profile could be a novel biomarker in AD. Here, we assessed the clotting profile in plasma from AD patients and age-matched controls. Utilizing clinically relevant assays, thromboelastography and activated partial thromboplastin time, we found impaired clot initiation and formation rate in AD patient plasma. These coagulation end points correlated with cerebrospinal fluid neurofilament-light levels and cognition and were more profound in younger AD patients. Ex vivo intrinsic clotting of plasma from AD mice expressing human amyloid precursor protein (APP) was also delayed in an age-dependent manner, suggesting that this phenotype is related to APP, the parent protein of Aß. Further analysis of coagulation factors in human plasma indicated that endogenous inhibitor(s) of factors XII and XI in AD plasma contribute to this delayed clotting. Together, these data suggest that delayed clotting in young AD patients is a novel biomarker and that therapies aimed to correct this phenotype might be beneficial in this patient population. Follow-up studies in additional AD patient cohorts are warranted to further evaluate these findings.


Asunto(s)
Enfermedad de Alzheimer/sangre , Precursor de Proteína beta-Amiloide/sangre , Inhibidores de Factor de Coagulación Sanguínea/sangre , Coagulación Sanguínea , Cognición , Factores de Edad , Anciano , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Biomarcadores/sangre , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Tiempo de Tromboplastina Parcial , Tromboelastografía
13.
Neurol Res ; 28(3): 250-5, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16687049

RESUMEN

The role of immune cells in promoting central nervous system (CNS) vascular permeability is poorly understood. In recent years, there is a growing body of literature that suggests CD8+ T-cells are potent mediators of vascular permeability in peripheral viral infections as well as in immune mediated neurological diseases. This review outlines the recent advances in tissue culture and animal models used to study vascular permeability. In addition, we put forth our hypothesis that CD8+ T-cells promote the opening of tight junctions between cerebral endothelial cells, enabling the infiltration of white blood cells and in certain models even leading to microhemorrhages in the CNS. Determining the mechanism by which CD8+ T-cells and other immune cells promote CNS vascular permeability in animal models could define new targets for immune mediated neurological conditions characterized by vascular permeability.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Permeabilidad Capilar/fisiología , Sistema Nervioso Central/citología , Animales , Humanos , Modelos Animales , Uniones Estrechas/fisiología
14.
PLoS One ; 8(3): e59032, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23516593

RESUMEN

The role of peripheral serotonin in nervous system development is poorly understood. Tryptophan hydroxylase-1 (TPH1) is expressed by non-neuronal cells including enterochromaffin cells of the gut, mast cells and the pineal gland and is the rate-limiting enzyme involved in the biosynthesis of peripheral serotonin. Serotonin released into circulation is taken up by platelets via the serotonin transporter and stored in dense granules. It has been previously reported that mouse embryos removed from Tph1-deficient mothers present abnormal nervous system morphology. The goal of this study was to assess whether Tph1-deficiency results in behavioral abnormalities. We did not find any differences between Tph1-deficient and wild-type mice in general motor behavior as tested by rotarod, grip-strength test, open field and beam walk. However, here we report that Tph1 (-/-) mice display altered gait dynamics and deficits in rearing behavior compared to wild-type (WT) suggesting that tryptophan hydroxylase-1 expression has an impact on the nervous system.


Asunto(s)
Marcha/genética , Triptófano Hidroxilasa/deficiencia , Animales , Electrocardiografía , Femenino , Marcha/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Serotonina/metabolismo , Triptófano Hidroxilasa/genética
15.
PLoS One ; 3(8): e3037, 2008 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-18725947

RESUMEN

Disruption of the blood brain barrier (BBB) is a hallmark feature of immune-mediated neurological disorders as diverse as viral hemorrhagic fevers, cerebral malaria and acute hemorrhagic leukoencephalitis. Although current models hypothesize that immune cells promote vascular permeability in human disease, the role CD8 T cells play in BBB breakdown remains poorly defined. Our laboratory has developed a novel murine model of CD8 T cell mediated central nervous system (CNS) vascular permeability using a variation of the Theiler's virus model of multiple sclerosis. In previous studies, we observed that MHC class II(-/-) (CD4 T cell deficient), IFN-gammaR(-/-), TNF-alpha(-/-), TNFR1(-/-), TNFR2(-/-), and TNFR1/TNFR2 double knockout mice as well as those with inhibition of IL-1 and LTbeta activity were susceptible to CNS vascular permeability. Therefore, the objective of this study was to determine the extent immune effector proteins utilized by CD8 T cells, perforin and FasL, contributed to CNS vascular permeability. Using techniques such as fluorescent activated cell sorting (FACS), T1 gadolinium-enhanced magnetic resonance imaging (MRI), FITC-albumin leakage assays, microvessel isolation, western blotting and immunofluorescent microscopy, we show that in vivo stimulation of CNS infiltrating antigen-specific CD8 T cells initiates astrocyte activation, alteration of BBB tight junction proteins and increased CNS vascular permeability in a non-apoptotic manner. Using the aforementioned techniques, we found that despite having similar expansion of CD8 T cells in the brain as wildtype and Fas Ligand deficient animals, perforin deficient mice were resistant to tight junction alterations and CNS vascular permeability. To our knowledge, this study is the first to demonstrate that CNS infiltrating antigen-specific CD8 T cells have the capacity to initiate BBB tight junction disruption through a non-apoptotic perforin dependent mechanism and our model is one of few that are useful for studies in this field. These novel findings are highly relevant to the development of therapies designed to control immune mediated CNS vascular permeability.


Asunto(s)
Barrera Hematoencefálica/inmunología , Linfocitos T CD8-positivos/inmunología , Animales , Astrocitos/fisiología , Barrera Hematoencefálica/fisiología , Sistema Nervioso Central/inmunología , Circulación Cerebrovascular/fisiología , Células Endoteliales/fisiología , Proteína Ligando Fas/deficiencia , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/fisiología , Perforina/fisiología , Proteínas Citotóxicas Formadoras de Poros/deficiencia , Linfocitos T Citotóxicos/inmunología , Uniones Estrechas/inmunología
16.
Int Rev Neurobiol ; 79: 73-97, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17531838

RESUMEN

Multiple sclerosis (MS) is the most common human demyelinating disease of the central nervous system. It is universally accepted that the immune system plays a major role in the pathogenesis of MS. For decades, CD4 T cells have been considered the predominant mediator of neuropathology in MS. This perception was largely due to the similarity between MS and CD4 T-cell-driven experimental allergic encephalomyelitis, the most commonly studied murine model of MS. Over the last decade, several new observations in MS research imply an emerging role for CD8 T cells in neuropathogenesis. In certain experimental autoimmune encephalomyelitis (EAE) models, CD8 T cells are considered suppressors of pathology, whereas in other EAE models, neuropathology can be exacerbated by adoptive transfer of CD8 T cells. Studies using the Theiler's murine encephalomyelitis virus (TMEV) model have demonstrated preservation of motor function and axonal integrity in animals deficient in CD8 T cells or their effector molecules. CD8 T cells have also been demonstrated to be important regulators of blood-brain barrier permeability. There is also an emerging role for CD8 T cells in human MS. Human genetic studies reveal an important role for HLA class I molecules in MS susceptibility. In addition, neuropathologic studies demonstrate that CD8 T cells are the most numerous inflammatory infiltrate in MS lesions at all stages of lesion development. CD8 T cells are also capable of damaging neurons and axons in vitro. In this chapter, we discuss the neuropathologic, genetic, and experimental evidence for a critical role of CD8 T cells in the pathogenesis of MS and its most frequently studied animal models. We also highlight important new avenues for future research.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Esclerosis Múltiple/patología , Esclerosis Múltiple/fisiopatología , Animales , Linfocitos T CD8-positivos/patología , Antígenos HLA-A/genética , Antígenos HLA-A/metabolismo , Antígeno HLA-A2 , Humanos , Modelos Biológicos , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Linfocitos T/clasificación , Linfocitos T/fisiología , Linfocitos T Reguladores/patología , Linfocitos T Reguladores/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA